WO2016020413A1 - Carrier molecule for antigens - Google Patents

Carrier molecule for antigens Download PDF

Info

Publication number
WO2016020413A1
WO2016020413A1 PCT/EP2015/068012 EP2015068012W WO2016020413A1 WO 2016020413 A1 WO2016020413 A1 WO 2016020413A1 EP 2015068012 W EP2015068012 W EP 2015068012W WO 2016020413 A1 WO2016020413 A1 WO 2016020413A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
polypeptide
acid sequence
saccharide
Prior art date
Application number
PCT/EP2015/068012
Other languages
English (en)
French (fr)
Inventor
Francesco Berti
Roberta COZZI
Guido Grandi
Domenico Maione
Immaculada Margarit Y Ros
Daniela Cira RINAUDO
Original Assignee
Glaxosmithkline Biologicals S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CA2957118A priority Critical patent/CA2957118A1/en
Priority to BR112017002183-8A priority patent/BR112017002183A2/pt
Priority to AU2015299008A priority patent/AU2015299008B2/en
Priority to SG11201700673RA priority patent/SG11201700673RA/en
Priority to KR1020177005840A priority patent/KR20170039709A/ko
Priority to US15/501,474 priority patent/US10245310B2/en
Priority to CN201580053997.XA priority patent/CN108064174A/zh
Priority to MX2017001638A priority patent/MX2017001638A/es
Application filed by Glaxosmithkline Biologicals S.A. filed Critical Glaxosmithkline Biologicals S.A.
Priority to JP2017506294A priority patent/JP2017523985A/ja
Priority to EP15744610.5A priority patent/EP3177324A1/en
Priority to EA201790239A priority patent/EA201790239A1/ru
Publication of WO2016020413A1 publication Critical patent/WO2016020413A1/en
Priority to IL250238A priority patent/IL250238A0/en
Priority to ZA2017/00824A priority patent/ZA201700824B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/095Neisseria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/315Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Streptococcus (G), e.g. Enterococci
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6068Other bacterial proteins, e.g. OMP

Definitions

  • the invention relates to conjugates of antigens and carrier molecules, and vaccines comprising these conjugates.
  • the antigens are typically saccharides.
  • TT tetanus toxoid
  • DT diphtheria toxoid
  • CRMi 97 diphtheria toxoid variant
  • the carrier proteins may be used in conjugates to induce protective and/or therapeutic immune responses against infections or drugs.
  • GBS Streptococcus agalactiae
  • BP-2a polypeptide a polypeptide having a spbl polypeptide
  • spbl polypeptide a polypeptide having a spbl polypeptide
  • These carriers are versatile and may be conjugated to various antigens, particularly saccharides e.g., from pathogenic organisms.
  • the resultant conjugates may be more immunogenic than conjugates based on currently used carrier proteins, e.g., CRMi 97 .
  • they may provide higher levels of protective immunity against GBS when used as carriers of GBS saccharides.
  • the invention therefore provides a conjugate comprising an antigen and a carrier molecule, wherein the carrier molecule comprises a BP-2a polypeptide and a spbl polypeptide.
  • the carrier molecule typically comprises the BP-2a polypeptide and a spbl polypeptide as a single polypeptide chain (a "hybrid" polypeptide).
  • the antigen is a saccharide.
  • the saccharide may be any saccharide, particularly a saccharide from a pathogenic organism.
  • the saccharide may be a capsular saccharide from Streptococcus agalactiae, Neisseria meningitidis, or Streptococcus pneumoniae, or a glucan.
  • the saccharide When the saccharide is a capsular saccharide from N. meningitidis, it is typically from one of the following meningococcal serogroups: A, C, W135 and Y. When the saccharide is a glucan, it is typically a laminarin. When the saccharide is a capsular saccharide from S. pneumoniae, it is typically from one of the following pneumococcal serotypes: 1 , 2, 3, 4, 5, 6A, 6B, 7F, 8, 9N, 9V, 10A, 1 1A, 12F, 14, 15B, 17F, 18C, 19A, 19F, 20, 22F, 23F and 33F.
  • polypeptide carriers of the invention as carriers for heterologous saccharides, i.e. for saccharides not found naturally in, for example Streptococcus agalactiae , may be advantageous in providing protection against two or more different microorganisms thereby extending vaccine coverage.
  • the present invention also relates to pharmaceutical compositions comprising a conjugate of the invention in combination with a pharmaceutically acceptable carrier.
  • the present invention relates to pharmaceutical compositions comprising (i) a conjugate according to the invention, (ii) saccharides from Streptococcus agalactiae serotype la, lb and III conjugated to CRM197 and (iii) a saccharide from Streptococcus agalactiae serotype II or V conjugated to GBS80.
  • the present invention relates to pharmaceutical compositions comprising (i) a saccharide from Streptococcus agalactiae serotype II conjugated to a GBS59(6XD3)-GBS 1523 carrier, (ii) saccharides from Streptococcus agalactiae serotype la, lb and III conjugated to CRM197 and (iii) a saccharide from Streptococcus agalactiae serotype V conjugated to GBS80.
  • the present invention further relates to methods for raising an immune response in a suitable mammal, comprising administering a conjugate or pharmaceutical composition of the invention to the mammal.
  • Figure 1 compares the protection (lower row of the table) and the anti-serotype-II saccharide antibody titres (graph and upper rows of the table) elicited by immunisation with conjugates comprising serotype-II saccharide, CRMi 97 and BP-2a/spbl carriers, in two studies. Compositions were administered at a dose of 1 ⁇ g protein.
  • Figure 2 displays the titre of antibodies specific for GBS59 and GBS 1523 elicited by the GBS59(6XD3)-GBS 1523-11 conjugate compositions, in two studies.
  • NB in this example the BP- 2a/spbl carrier is referred to as GBS59(6XD3)-GBS1523.
  • Figure 3 compares the anti-serotype-II saccharide antibody titres elicited by immunisation with conjugates comprising serotype-II saccharide and CRM197, GBS80 and BP-2a/spbl carriers.
  • Figure 4 compares the anti-type-V saccharide antibody titres elicited by immunisation with conjugates comprising serotype-V saccharide and CRM197, GBS80 and BP-2a/spbl carriers.
  • Figure 5 compares the protection (lower rows of the table) and the anti-type-V saccharide antibody titres (graph and upper row of the table) elicited by immunisation with conjugates comprising serotype-V saccharide and CRMi 97 and BP-2a/spbl carriers. Compositions were administered at a dose of 1 ⁇ g protein.
  • Figure 6 displays the titre of antibodies specific for GBS59 and GBS 1523 elicited by the GBS59(6XD3)-GBS1523-V conjugate compositions, in two studies.
  • NB in this example the BP- 2a/spbl carrier is referred to as GBS59(6XD3)-GBS1523.
  • Figure 8 compares the anti-type-II saccharide antibody titres elicited by immunisation with various mixtures of conjugates.
  • Figure 9 compares the anti-type-V saccharide antibody titres elicited by immunisation with various mixtures of conjugates.
  • Figure 10 presents some of the data of Figure 8 and indicates certain statistically significant differences in antibody titres.
  • Figure 11 presents some of the data of Figure 9 and indicates certain statistically significant differences in antibody titres.
  • Figure 12 provides the sequence of carrier protein GBS59(6XD3)-GBS 1523 (SEQ ID NO:276) and highlights its constituent portions. Linker sequences are boxed, whilst different BP-2a and spbl sequences are marked with alternating single and double underlining.
  • Figure 13 provides the repeating units of representative bacterial saccharides for use in the conjugates of the invention.
  • Figure 14 presents western blotting with serum from human patient which strongly recognizes full length spbl (GBS 1523) and spbl D2+D 3 (GBS 1523 D2+D 3) but not spbl D1 (GBS 1523 D1 ).
  • the invention involves a conjugate comprising an antigen and a carrier molecule, wherein the carrier molecule comprises a BP-2a polypeptide and a spbl polypeptide.
  • the carrier molecule comprises a BP-2a polypeptide and a spbl polypeptide.
  • GBS protein antigens particularly as carriers of polysaccharide, is advantageous because unlike non-GBS carriers, such as CRM197, GBS protein antigens elicit a protective immunologic response against GBS.
  • GBS carrier molecules of the invention as protein carriers for GBS polysaccharide(s) in a vaccine composition increases vaccine coverage.
  • Non-prevalent serotypes of colonizing GBS may emerge in a population vaccinated with a multivalent vaccine against currently predominant colonizing serotypes in a process called "serotype replacement".
  • serotype replacement a multivalent vaccine against currently predominant colonizing serotypes in a process called "serotype replacement".
  • conjugates of the invention may also tackle such concerns around serotype replacement.
  • the carrier molecule comprises a BP-2a polypeptide and a spbl polypeptide.
  • the carrier molecule comprises the BP-2a polypeptide and the spbl polypeptide as a single polypeptide chain (a "hybrid" polypeptide).
  • Streptococcus agalactiae (Group B Streptococcus, GBS) has three pilus variants, each encoded by a distinct pathogenicity island, PI-1, PI-2a and PI-2b [18,19].
  • Each pathogenicity island consists of 5 genes coding for: the pilus backbone protein (BP); 2 ancillary proteins (API and AP2); and 2 sortase proteins that are involved in the assembly of the pili. All GBS strains carry at least one of these 3 pathogenicity islands and the sequences of the pilus structural proteins (BP, API and AP2) encoded by these pathogenicity islands are generally well conserved.
  • the sequence of the backbone protein encoded by pathogenicity island 2a varies between GBS strains.
  • the BP-2a pilus subunit has at least seven clades and the sequence identity between these clades is as low as 48%.
  • the BP-2a polypeptide may also be referred to as GBS59 or SAL1486.
  • Reference amino acid sequences for the seven BP-2a clades are SEQ ID NO: l (derived from GBS strain 2603), SEQ ID NO:2 (derived from GBS strain 515), SEQ ID NO:3 (derived from GBS strain CJB111), SEQ ID NO:4 (derived from GBS strain H36B), SEQ ID NO:5 (derived from GBS strain CJB110), SEQ ID NO:6 (derived from GBS strain DK21) and SEQ ID NO:7 (derived from GBS strain NEM316) herein.
  • the inventors have determined that BP-2a polypeptides are useful as carrier molecules when combined with a spbl polypeptide.
  • Preferred BP-2a polypeptides for use with the invention comprise: an amino acid sequence selected from the group consisting of SEQ ID NOs: l-7; a fragment of at least 20 contiguous amino acids (e.g. at least 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250) of an amino acid sequence selected from the group consisting of SEQ ID NOs: l-7; or an amino acid sequence having at least 70% identity (e.g,.
  • the BP-2a polypeptide of the invention preferably comprises at least one CD4+ T cell epitope.
  • Preferred fragments lack one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or more) from the C-terminus and/or one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or more) from the N-terminus of an amino acid sequence selected from the group consisting of SEQ ID NOs: l-7 while retaining at least one epitope of an amino acid sequence selected from the group consisting of SEQ ID NOs: l-7.
  • Other fragments omit one or more protein domains.
  • the BP-2a polypeptide of the invention comprises at least one CD4 + T cell epitope.
  • CD4 + T cells help B lymphocytes to produce antibodies against antigens [20].
  • T-cell epitopes can be identified empirically (e.g. using PEPSCAN [21 ,22] or similar methods), or they can be predicted (e.g. using the Jameson-Wolf antigenic index [23], matrix-based approaches [24], TEPITOPE [25], neural networks [26], OptiMer & EpiMer [27,28], ADEPT [29], Tsites [30], hydrophilicity [31], antigenic index [32] or the methods disclosed in reference 33, etc.).
  • Preferred BP-2a polypeptides for use with the invention comprise or consist of immunogenic domains that may comprise important CD4+ T cell epitopes. Exemplary domains present in the different BP-2a clades are provided below.
  • the BP-2a protein can be split into four domains (D 1 to D4) between the end of its leader peptide and the start of its LPXTG anchor. These four domains are as follows in SEQ ID NOs: 1 to 7, and the positions in further BP-2a sequences which correspond to these residues can readily be identified by alignment:
  • the sub-fragments of domain D3 identified below are surface-exposed fragments.
  • the inventors have identified that these D3 sub-fragments may be particularly effective carriers when combined with spbl polypeptides.
  • BP-2a polypeptides for use with the invention comprise an amino acid sequence selected from the above domains, a fragment of at least 20 amino acids of one of the above domains, or an amino acid sequence having at least 70% identity to said domains or fragments.
  • the BP-2a polypeptides for use with the invention comprise a D3 fragment, of homologue thereof. D3 fragments have been shown to induce a protective immune response [34].
  • the BP-2a polypeptides for use with the invention comprise or consist of a D3 sub-fragment. Such polypeptides may be particularly effective carriers when used in combination with an spbl polypeptide.
  • BP-2a polypeptides for use with the invention may consist of fragments of SEQ ID NOS: 1 , 2, 3, 4, 5, 6 or 7 comprising or consisting of domain D3 or a D3 sub-fragment, as identified above.
  • Particularly preferred BP-2a polypeptides for use with the invention comprise: an amino acid sequence selected from the group consisting of SEQ ID NOs: 36, 38, 40, 42, 44, 46 and 48; a fragment of at least 20 contiguous amino acids (e.g., at least 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250) of an amino acid sequence selected from the group consisting of SEQ ID NOs: 36, 38, 40, 42, 44, 46 and 48; or an amino acid sequence having at least 70% identity (e.g., at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%), 96%), 97%o, 98%o, 99%o, 99.5%o) to an amino acid
  • BP-2a polypeptides for use with the invention comprise: a fragment comprising or consisting of no more than 20 contiguous amino acids (for example, no more than 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 contiguous amino acids) of an amino acid sequence selected from the group consisting of SEQ ID NOs: 36, 38, 40, 42, 44, 46 and 48.
  • Fragments of the D3 sub-fragments are envisaged, for example, fragments lacking one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or more) from the C-terminus and/or one or more amino acids (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or more) from the N-terminus of an amino acid sequence selected from the group consisting of SEQ ID NOs: 36, 38, 40, 42, 44, 46 and 48 while retaining at least one epitope of an amino acid sequence selected from the group consisting of SEQ ID NOs: 36, 38, 40, 42, 44, 46 and 48.
  • a polypeptide may consist of a fragment of SEQ ID NO:3 comprising SEQ ID NO:78 (amino acids 41 1 to 436 of SEQ ID NO:3).
  • fragments may retain domains D4, D2 and/or Dl in addition to epitopes from domain D3.
  • Polypeptides of the invention may therefore consist of fragments of SEQ ID NOS: 1 , 2, 3, 4, 5, 6 or 7 comprising or consisting of i) domains D2 and D3; ii) domains D3 and D4; iii) domains Dl , D2 and D3; or iv) domains D2, D3 and D4, as identified above.
  • Exemplary fragments are provided below. These fragments containing combinations of domains D2, D3, and D4 (or D4H) as set out below:
  • SEQ ID NO:4 (h36b) SEQ ID NO: 62 SEQ ID NO: 63 SEQ ID NO: 64 SEQ ID NO: 65
  • SEQ ID NO:5 CJB110
  • SEQ ID NO: 66 SEQ ID NO: 67
  • SEQ ID NO:68 SEQ ID NO: 69
  • SEQ ID NO: 6 (DK21) SEQ ID NO: 70 SEQ ID NO:71 SEQ ID NO: 72 SEQ ID NO: 73
  • SEQ ID NO: 7 SEQ ID NO: 74 SEQ ID NO: 75 SEQ ID NO: 76 SEQ ID NO: 77 (NEM316)
  • Amino acid sequences used with the invention may, compared to SEQ ID NOs: 1 , 2, 3, 4, 5, 6, or 7 or fragments thereof include one or more (e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) conservative amino acid replacements i. e. replacements of one amino acid with another which has a related side chain.
  • Genetically-encoded amino acids are generally divided into four families: (1) acidic i.e. aspartate, glutamate; (2) basic i.e. lysine, arginine, histidine; (3) non-polar i.e.
  • the polypeptides may have one or more (e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) single amino acid deletions relative to a reference sequence.
  • the polypeptides may also include one or more (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) insertions (e.g. each of 1 , 2, 3, 4 or 5 amino acids) relative to a reference sequence.
  • the carrier comprises more than one BP-2a polypeptide.
  • the inventors have shown that carriers comprising multiple BP-2a polypeptides may be particularly effective carriers when used in combination with an spbl polypeptide. Therefore, in preferred embodiments, the carrier comprises at least 2, at least 3, at least 4, at least 5, at least 6 or at least 7 BP-2a polypeptides. When more than one BP-2a polypeptide is present, each BP-2a polypeptide is generally from a different strain.
  • Conjugates of the invention may comprise: at least two (e.g. at least 3, at least 4, at least 5, at least 6 or at least 7) amino acid sequences selected from the group consisting of SEQ ID NOs: 1-7; at least two fragments of at least 20 contiguous amino acids (e.g. 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more) or no more than 20 contiguous amino acids (e.g. 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or fewer) of amino acid sequences selected from the group consisting of SEQ ID NOs: 1-7; or at least two amino acid sequences having at least 70% identity (e.g.
  • amino acid sequences selected from the group consisting of SEQ ID NOs: 1-7 or having said identity to said fragments of amino acid sequences selected from the group consisting of SEQ ID NOs: 1-7.
  • the carrier may comprise corresponding BP-2a domains from different strains.
  • the carrier may comprise D3 domains or D3 sub-fragments from 2, 3, 4, 5, 6 or 7 different strains.
  • each BP-2a polypeptide will be present in a single polypeptide chain.
  • linker amino acid sequences will be apparent to those skilled in the art.
  • useful linkers may include GSGS (SEQ ID NO:277), GSGGGG (SEQ ID NO:272) or GSGSGGGG (SEQ ID NO:273).
  • the polypeptide chain comprising BP-2a polypeptides (and optionally linkers) can be expressed as a fusion polypeptide using standard recombination expression techniques.
  • single polypeptide chain generally refers to a polypeptide chain in which all amino acids are linked sequentially by peptide bonds.
  • a "single polypeptide chain” is usually generated by expression of a single DNA sequence.
  • two or more BP-2a polypeptides may be linked using standard chemical conjugation techniques, e.g., using straight or branched-chain carbon linkers, heterocyclic carbon linkers, carbohydrate linkers.
  • a linker can be attached through lysine or cysteine side chains.
  • recombinant fusion protein expression and chemical conjugation techniques may be used in combination to produce a single polypeptide chain comprising multiple BP-2a polypeptides.
  • the different BP-2a polypeptides may be present in any order in the polypeptide chain. Exemplary combinations are provided below. Each number in the table indicates the relevant SEQ ID NO.
  • examples of BP-2a polypeptide hybrids suitable for use in the invention include polypeptides comprising an amino acid sequence selected from the group consisting of: SEQ ID NO: 83; SEQ ID NO: 84; SEQ ID NO: 85; SEQ ID NO: 86; or SEQ ID NO: 87, and homologues having at least 70% identity to said sequences ⁇ e.g. at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%).
  • Other suitable BP-2a hybrids are disclosed in International patent application publication WO201 1/121576. spbl
  • spbl The original spbl (SAN1518; GBS1523) sequence was annotated in reference 35 as a cell wall surface anchor family protein (see GI: 77408651 or Uniprot accession No. Q8E479).
  • the terms 'spbl ' and 'GBS 1523 ' are used interchangeably herein.
  • amino acid sequence of full length spbl as found in the COH1 strain is given as SEQ ID NO: 206 herein.
  • Preferred spbl polypeptides for use with the invention comprise: the amino acid sequence of SEQ ID NO:206; a fragment of at least 20 contiguous amino acids ⁇ e.g.
  • amino acid sequence of SEQ ID NO:206 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more) of the amino acid sequence of SEQ ID NO:206; a fragment of less than 400 contiguous amino acids (e.g. 395, 390, 389, 388, 387, 386, 385, 384, 383 or less) of the amino acid sequence of SEQ ID NO:206; or an amino acid sequence having at least 70% identity (e.g. at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%) to the amino acid sequence of SEQ ID NO:206 or having said identity to said fragment of the amino acid sequence of SEQ ID NO:206.
  • 70% identity e.g. at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%
  • Particular spbl fragments of the invention include fragment spb l D2+D3 (referred to in the Examples as BP-2bD2+D3 or BP-2b i85-46s), a fragment consisting of amino acids 185 to 468 of full length spb l , particularly amino acids 185 to 468 of SEQ ID NO: 206 provided as SEQ ID NO:285.
  • the inventors have shown that spbl polypeptides may be particularly effective carriers when used in combination with a BP-2a polypeptide.
  • These spbl proteins include variants of SEQ ID NO: 206.
  • the spbl polypeptide of the invention preferably comprises at least one CD4+ T cell epitope.
  • Preferred fragments lack one or more amino acids (e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or more) from the C-terminus and/or one or more amino acids (e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or more) from the N-terminus of SEQ ID NO: 206 while retaining at least one epitope of SEQ ID NO: 206.
  • Other fragments omit one or more protein domains.
  • Wild-type spb l contains a N-terminal leader or signal sequence region at amino acids 1 to 30 of SEQ ID NO:206 which may be removed in fragments, e.g. SEQ ID NO:207.
  • amino acids 468-502 of SEQ ID NO:206 may be removed in fragments, e.g. SEQ ID NO:207.
  • amino acids 1-185 of SEQ ID NO:206 may be removed in fragments. Therefore, in preferred embodiments, spbl polypeptides for use with the invention comprise: the amino acid sequence of SEQ ID NO:207; a fragment of at least 20 contiguous amino acids (e.g. 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250 or more) of the amino acid sequence of SEQ ID NO:207; a fragment of no more than 20 contiguous amino acids (e.g.
  • the spbl polypeptide comprises or consists of the amino acid sequence of SEQ ID NO:207.
  • the wild-type spbl sequence contains an amino acid motif indicative of a cell wall anchor (LPSTG) at amino acids 468-472 of SEQ ID NO:206.
  • LSTG cell wall anchor
  • the extracellular domain of the expressed polypeptide may be cleaved during purification or the recombinant polypeptide may be left attached to either inactivated host cells or cell membranes in the final composition.
  • An E box containing a conserved glutamic residue has also been identified at amino acids 419-429 of SEQ ID NO:206, with a conserved glutamic acid at residue 423.
  • the E box motif may be important for the formation of oligomeric pilus-like structures, and so useful fragments of spbl may include the conserved glutamic acid residue.
  • a mutant of spbl has been identified in which the glutamine (Q) at position 41 of SEQ ID NO:206 is substituted for a lysine (K), as a result of a mutation of a codon in the encoding nucleotide sequence from CAA to AAA. This substitution may be present in the spb l sequences and spbl fragments (e.g. SEQ ID NO:208).
  • Hybrid polypeptides can be represented by the formula NH 2 -A- ⁇ -X-L- ⁇ diligent-B-COOH, wherein: A is an optional N-terminal amino acid sequence; B is an optional C-terminal amino acid sequence; n is an integer of 2 or more (e.g.
  • each X is an amino acid sequence of a spbl polypeptide or a BP-2a polypeptide (as described above), wherein at least one X is a spbl polypeptide and at least one X is a BP-2a polypeptide; and L is an optional linker amino acid sequence.
  • n 2
  • Xi is usually a BP-2a polypeptide
  • X 2 is usually a spbl polypeptide.
  • each spbl polypeptide (when more than one is present) may be the same or different and each BP-2a polypeptide (when more than one is present) may be the same or different.
  • the spbl polypeptide or BP-2a polypeptide that is the amino acid sequence of each X is as defined above. Where these polypeptides are defined in terms of comprising an amino acid sequence having at least a certain percentage identity to a given spbl or BP-2a polypeptide or fragment, the level of identity may be the same for each X.
  • linker amino acid sequence -L- may be present or absent.
  • the hybrid may be NHz-Xi-Lj-Xz-Lz-COOH, NH 2 -Xi-X 2 -COOH, NH 2 -Xi-Li-X 2 - COOH, NH 2 -X 1 -X 2 -L 2 -COOH, etc.
  • Linker amino acid sequence(s) -L- will typically be short (e.g. 20 or fewer amino acids i.e. 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2, 1).
  • Other suitable linker amino acid sequences will be apparent to those skilled in the art.
  • a useful linker is GSGS (SEQ ID NO:277), GSGGGG (SEQ ID NO:272) or GSGSGGGG (SEQ ID NO:273), with the Gly-Ser dipeptide being formed from a BamHl restriction site, thus aiding cloning and manipulation, and the (Gly) 4 tetrapeptide (SEQ ID NO: 297) being a typical poly-glycine linker.
  • Other suitable linkers, particularly for use as the final L n are a Leu-Glu dipeptide or SEQ ID NO:274.
  • SEQ ID NO:277 is preferred for linking different BP-2a polypeptides.
  • SEQ ID NO:272 is preferred for linking BP-2a polypeptides and spbl polypeptides.
  • -A- is an optional N-terminal amino acid sequence.
  • This will typically be short (e.g. 40 or fewer amino acids i.e. 40, 39, 38, 37, 36, 35, 34, 33, 32, 31 , 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2, 1).
  • Other suitable N-terminal amino acid sequences will be apparent to those skilled in the art.
  • -A- is preferably an oligopeptide (e.g. with 1 , 2, 3, 4, 5, 6, 7 or 8 amino acids) which provides a N-terminus methionine e.g. Met-Ala-Ser, or a single Met residue.
  • a preferred -A- is a single Met residue.
  • -A-, -B- and -L- sequences do not include a sequence that shares 10 or more contiguous amino acids in common with a human polypeptide sequence.
  • Particular carrier molecules may comprise a fusion of (i) at least one D3-sub fragment selected from the group consisting of GBS strains 515, H36B, CJB 1 1 1 , 2603, CJB 1 10 and DK21 and (ii) an spbl polypeptide or fragment thereof. More particularly carrier molecules may comprise a fusion of (i) at least one D3 sub-fragment sequence selected from the group consisting of SEQ ID NO:38, SEQ ID NO:42, SEQ ID NO:40, SEQ ID NO:36, SEQ ID NO:44and SEQ ID NO:46 and (ii) an spbl polypeptide of SEQ ID NO:207.
  • carrier molecules may comprise a fusion of (i) at least one D3 sub-fragment sequence selected from the group consisting of SEQ ID NO:38, SEQ ID NO:42, SEQ ID NO:40, SEQ ID NO:36, SEQ ID NO:44and SEQ ID NO:46 and (ii) a D2+D3 fragment of the spbl polypeptide, particularly a fragment comprising or consisting of amino acids 185 to 468 of SEQ ID NO:206.
  • an exemplary hybrid is carrier GBS59(6XD3)-GBS 1523, presented in SEQ ID NO:276.
  • the carrier molecule of the invention comprises an amino acid sequence having at least 70% identity (e.g. at least 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%) to SEQ ID NO:276 (as determined across the length of SEQ ID NO:276).
  • the carrier molecule comprises at least one spbl CD4 + T cell epitope and at least one BP-2a CD4 + T cell epitope from each of BP-2a (515), BP-2a (H36B), BP-2a (CJB l l l), BP-2a (2603), BP-2a (CJB 1 10) and BP-2a (DK21).
  • SEQ ID NO:276 The amino acid sequence of SEQ ID NO:276 is shown schematically in Figure 12.
  • This particular carrier molecule consists of the following parts from N-terminus to C-terminus: N-terminal methionine; D3-sub fragment from 515 (SEQ ID NO:38); GSGS linker (SEQ ID NO:277); D3-sub fragment from H36B (SEQ ID NO:42); GSGS linker (SEQ ID NO:277); D3-sub fragment from CJB l l l (SEQ ID NO:40); GSGS linker (SEQ ID NO:277); D3-sub fragment from 2603 (SEQ ID NO:36); GSGS linker (SEQ ID NO:277); D3-sub fragment from CJB 1 10 (SEQ ID NO:44); GSGS linker (SEQ ID NO:277); D3-sub fragment from DK21 (SEQ ID NO:46); GSGGGG linker (SEQ ID NO:272); s
  • the carrier comprises at least 1 (i.e. 1 , 2, 3, 4, 5, 6, 7 or more) BP-2a polypeptides from different strains.
  • the carrier comprises at least 2 BP-2a polypeptides. Even more preferably, the carrier comprises at least 4, 5, 6 or 7 BP-2a polypeptides from different strains.
  • the BP-2a and spb 1 polypeptides may be present in the carrier molecule in any order.
  • the spbl polypeptide is towards or is at the C-terminus (i.e. the final -X- is an spbl polypeptide).
  • all of the BP-2a polypeptides present in the carrier are grouped together and are all either N-terminal or C-terminal to the spbl polypeptide.
  • the hybrid polypeptide can be represented by the formula NH2-A- ⁇ -X B p-2a-L- ⁇ till-X S pbi-L-B-COOFI, wherein n is at least 1 , and is preferably at least 2, and each Xep-2a is derived from a different strain.
  • the carrier may comprise other polypeptides in addition to the spb l and BP-2a polypeptides.
  • the carrier may comprise GBS80 and/or GBS67 polypeptides, or homologues or fragments thereof.
  • the amino acid sequence of GBS80 and GBS67 polypeptides are provided in SEQ ID NOs: 177-205.
  • GBS80 is also known as SAG0645 identified in Uniprot under Accession No. Q8E0S9 (Cell wall surface anchor family protein).
  • Hybrid polypeptide may be produced using recombinant fusion protein expression methods, chemical conjugation techniques, or any combination, as described above.
  • Carrier molecules modified to incorporate non-natural amino acids are Carrier molecules modified to incorporate non-natural amino acids
  • the invention also involves carrier molecules which have been modified to incorporate non-natural amino acids.
  • the non-natural amino acid may be used to conjugate the carrier molecule to another molecule.
  • the carrier molecule comprises one or more (e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 etc.) non-natural amino acids.
  • the non-natural amino acid may have a functional group with a reaction profile that is different to the functional groups available to react in proteins composed of the canonical amino acids (e.g. the amino group of lysine or the sulhydryl group of cysteine). This in turn means that chemoselective reactions allow site-selective conjugations to be performed at pre-determined sites where a non-natural amino acid has been incorporated into the protein.
  • the carrier molecule may comprise one or more L-homoallylglycine (HAG) residues.
  • HAG residues are substituted in place of the methionine residues in the sequence.
  • HAG chemically known as L-2-amino-5-hexenoic acid, is an analogue of methionine, and contains a reactive alkene site.
  • HAG can substitute for methionine in both the initiation and elongation steps of protein synthesis.
  • HAG has an olefinic side-chain which has a different reaction profile to the functional groups found in canonical amino acids, reacting through a thiyl-ene mechanism.
  • the carrier molecule may be modified to include other non-natural amino acids which permit site-selective conjugations to be performed at pre-determined sites.
  • the carrier molecule may be modified so that one or more (e.g. 1 , 2, 3, 4, 5, etc.) p- acetylphenylalanine residues are included in its sequence.
  • This amino acid has a keto functional group, which is not present in any of the canonical amino acids, and therefore the amino acid can be reacted specifically with hydrazines, alkoxyamines and semicarbazides under mild aqueous conditions to produce hydrazone, oxime and semicarbazone linkages.
  • Other amino acids with keto functional groups include m-acetylphenylalanine and / benzoylphenylalanine and these residues may be used in the same manner.
  • the carrier molecule may be modified to include an azide group (which also does not occur in the canonical amino acids), for example by incorporation of one or more (e.g. 1 , 2, 3, 4, 5, etc.) -azidophenylalanine residues.
  • the azide group can react with an acetylene group on the conjugation partner through a copper (I) catalysed [2+3] cycloaddition reaction.
  • the carrier molecule may be modified to include one or more (e.g. 1 , 2, 3, 4, 5, etc.) phenylselenocysteine residues. Treatment of this residue with hydrogen peroxide allows its conjugation to thiol groups.
  • the antigen of the antigen is the antigen
  • the antigen is typically a saccharide.
  • the saccharide may be any saccharide, particularly a saccharide from a pathogenic organism. Exemplary saccharides for use in the invention are described below.
  • the saccharide may be a bacterial saccharide, e.g. a bacterial capsular saccharide. Representative bacterial saccharides are described in Figure 13.
  • the saccharides may be used in the form of oligosaccharides. These are conveniently formed by fragmentation of purified polysaccharide (e.g. by hydrolysis), which will usually be followed by purification of the fragments of the desired size. Saccharides may be purified from natural sources. Fragmentation of polysaccharides is typically performed to give a final average degree of polymerisation (DP) in the oligosaccharide of less than 30 (e.g. between 10 and 20, around 10 (for example, for serogroup A); between 15 and 25 (for example, for serogroups W135 and Y), around 15-20; between 12 and 22 (for example, for serogroup C); etc.). DP can conveniently be measured by ion exchange chromatography or by colorimetric assays [36].
  • DP can conveniently be measured by ion exchange chromatography or by colorimetric assays [36].
  • the hydrolysate will generally be sized in order to remove short-length oligosaccharides [37]. This can be achieved in various ways, such as ultrafiltration followed by ion-exchange chromatography. Oligosaccharides with a degree of polymerisation of less than or equal to about 6 are preferably removed (for example, for serogroup A), and those less than around 4 are preferably removed (for example, for serogroups W135 and Y).
  • Chemical hydrolysis of saccharides generally involves treatment with either acid or base under conditions that are standard in the art. Conditions for depolymerisation of capsular saccharides to their constituent monosaccharides are known in the art.
  • One depolymerisation method involves the use of hydrogen peroxide [38]. Hydrogen peroxide is added to a saccharide (e.g. to give a final H 2 O 2 concentration of 1%), and the mixture is then incubated (e.g. at around 55°C) until a desired chain length reduction has been achieved. The reduction over time can be followed by removing samples from the mixture and then measuring the (average) molecular size of saccharide in the sample. Depolymerization can then be stopped by rapid cooling once a desired chain length has been reached.
  • saccharides may be obtained by total or partial synthesis.
  • the antigen when it is not a saccharide, it may be any other antigen, i.e. any immunogen or hapten. Conjugates of the invention may elicit an immune response against a hapten conjugated to the carrier molecule.
  • the hapten may for example be a drug of abuse [39].
  • Examples include, but are not limited to, opiates, marijuana, amphetamines, cocaine, barbituates, glutethimide, methyprylon, chloral hydrate, methaqualone, benzodiazepines, LSD, nicotine, anticholinergic drugs, antipsychotic drugs, tryptamine, other psychomimetic drugs, sedatives, phencyclidine, psilocybine, volatile nitrite, and other drugs inducing physical and/or psychological dependence.
  • Preferred exemplary bacterial capsular saccharides include those from Streptococcus agalactiae.
  • the Examples demonstrate that carriers comprising BP-2a polypeptides and spbl polypeptides may be effective carriers for GBS saccharides.
  • carriers comprising BP-2a polypeptides and spbl polypeptides may induce carrier-specific immune responses that provide useful protection.
  • the GBS capsular saccharide is covalently linked to the peptidoglycan backbone of GBS, and is distinct from the group B antigen, which is another saccharide that is attached to the peptidoglycan backbone.
  • the GBS capsular saccharides are chemically related, and all GBS capsular saccharides share the following trisaccharide core:
  • the various GBS serotypes differ by the way in which this core is modified.
  • the difference between serotypes la and III arises from the use of either the GlcNAc (la) or the Gal (III) in this core for linking consecutive trisaccharide cores.
  • Serotypes la and lb both have a [a-D-Neu/?NAc(2 ⁇ 3)P-D-Gal/ (l ⁇ ] disaccharide linked to the GlcNAc in the core, but the linkage is either 1 ⁇ 4 (la) or 1 ⁇ 3 (lb).
  • GBS-related disease arises primarily from serotypes la, lb, II, III, IV, V, VI, VII, and VIII, with over 85% being caused by five serotypes: la, lb, II, III & V.
  • the invention may use a saccharide from one or more of these eight serotypes, particularly from one or more of the serotypes selected from the group consisting of la, lb, II, III, IV and V, yet more particularly one or more polysaccharides selected from the group consisting of serotypes la, lb, II, III and V.
  • the capsular saccharides of each of these serotypes include: (a) a terminal N-acetyl-neuraminic acid (NeuNAc) residue (commonly referred to as sialic acid), which in all cases is linked 2 ⁇ 3 to a galactose residue; and (b) a N-acetyl- glucosamine residue (GlcNAc) within the trisaccharide core.
  • NeuroNAc N-acetyl-neuraminic acid
  • sialic acid commonly referred to as sialic acid
  • GlcNAc N-acetyl- glucosamine residue
  • All five saccharides include galactose residues within the trisaccharide core, but serotypes la, lb, II & III also contain additional galactose residues in each repeating unit.
  • the conjugate of the invention comprises a serotype II capsular saccharide or a serotype V capsular saccharide.
  • the Examples demonstrate that the BP-2a/spbl carriers of the invention may be particularly effective carriers for serotype II capsular saccharide and serotype V, and may be more effective than CRM.
  • the conjugate of the invention comprises a serotype II capsular saccharide.
  • the Examples demonstrate that the BP-2a/spbl carriers of the invention may be particularly effective carriers for serotype II capsular saccharide.
  • Saccharides used according to the invention may be in their native form, or may have been modified. For example, the saccharide may be shorter than the native capsular saccharide, or may be chemically modified.
  • the serotype V capsular saccharide used in the invention may be modified as described in refs. 40 and 41.
  • a serotype V capsular saccharide that has been substantially desialylated may be prepared by treating purified GBS serotype V capsular saccharide under mildly acidic conditions (e.g. 0.1M sulphuric acid at 80°C for 60 minutes) or by treatment with neuraminidase, as described in reference 40.
  • the saccharide used according to the invention may be a substantially full-length capsular polysaccharide, as found in nature, or it may be shorter than the natural length.
  • Full-length polysaccharides may be depolymerised to give shorter fragments for use with the invention e.g. by hydrolysis in mild acid, by heating, by sizing chromatography, etc.
  • the serotype II and/or III capsular saccharides used in the invention may be depolymerised as described in refs. 42 and 43.
  • the saccharide may be chemically modified relative to the capsular saccharide as found in nature.
  • the saccharide may be de-O-acetylated (partially or fully), de-N-acetylated (partially or fully), N-propionated (partially or fully), etc.
  • De-acetylation may occur before, during or after conjugation, but preferably occurs before conjugation.
  • de-acetylation may or may not affect immunogenicity.
  • O-acetylation on GBS saccharides in various serotypes is discussed in reference 44, and in some embodiments O-acetylation of sialic acid residues at positions 7, 8 and/or 9 is retained before, during and after conjugation e.g.
  • the GBS saccharide used in the present invention has substantially no O-acetylation of sialic acid residues at positions 7, 8 and/or 9.
  • O-acetylation is typically lost. The effect of de-acetylation etc. can be assessed by routine assays.
  • Capsular saccharides can be purified by known techniques, as described in ref. 45.
  • a typical process involves base extraction, centrifugation, filtration, RNase/DNase treatment, protease treatment, concentration, size exclusion chromatography, ultrafiltration, anion exchange chromatography, and further ultrafiltration.
  • Treatment of GBS cells with the enzyme mutanolysin, which cleaves the bacterial cell wall to free the cell wall components, is also useful.
  • the purification process described in reference 46 can be used. This involves base extraction, ethanol/CaCl 2 treatment, CTAB precipitation, and re-solubilisation. A further alternative process is described in ref. 47.
  • Exemplary capsular saccharides for use with the carriers of the invention include those from
  • N. meningitidis have been identified, including A, B, C, H, I, K, L, 29E, W135, X, Y & Z.
  • the saccharide in the invention may be from any of these serogroups.
  • the saccharide is from one of the following meningococcal serogroups: A, C, W135 and Y.
  • a more preferred process [49] involves polysaccharide precipitation followed by solubilisation of the precipitated polysaccharide using a lower alcohol.
  • Precipitation can be achieved using a cationic detergent such as tetrabutylammonium and cetyltrimethylammonium salts (e.g. the bromide salts), or hexadimethrine bromide and myristyltrimethylammonium salts.
  • cetyltrimethylammonium bromide 'CTAB' is particularly preferred [50].
  • Solubilisation of the precipitated material can be achieved using a lower alcohol such as methanol, propan-l-ol, propan-2-ol, butan-l-ol, butan-2-ol, 2-methyl- propan-l-ol, 2-methyl-propan-2-ol, diols, etc., but ethanol is particularly suitable for solubihsing CTAB-polysaccharide complexes.
  • Ethanol may be added to the precipitated polysaccharide to give a final ethanol concentration (based on total content of ethanol and water) of between 50% and 95%.
  • the polysaccharide may be further treated to remove contaminants. This is particularly important in situations where even minor contamination is not acceptable (e.g. for human vaccine production). This will typically involve one or more steps of filtration e.g. depth filtration, filtration through activated carbon may be used, size filtration and/or ultrafiltration. Once filtered to remove contaminants, the polysaccharide may be precipitated for further treatment and/or processing. This can be conveniently achieved by exchanging cations (e.g. by the addition of calcium or sodium salts).
  • the capsular saccharides are conjugated to carrier proteins as described below. Further and alternative methods for purification and conjugation of meningococcal saccharides are disclosed in refs. 38 & 51. As an alternative to purification, capsular saccharides of the present invention may be obtained by total or partial synthesis e.g. Hib synthesis is disclosed in ref. 52, and MenA synthesis in ref. 53.
  • the saccharide may be chemically modified e.g. it may be O-acetylated or de-O-acetylated. Any such de-O-acetylation or hyper-acetylation may be at specific positions in the saccharide. For instance, most serogroup C strains have O-acetyl groups at position C-7 and/or C-8 of the sialic acid residues, but about 15% of clinical isolates lack these O-acetyl groups [54,55]. The acetylation does not seem to affect protective efficacy (e.g. unlike the MenjugateTM product, the NeisVac-CTM product uses a de-O-acetylated saccharide, but both vaccines are effective).
  • the serogroup W135 saccharide is a polymer of sialic acid-galactose disaccharide units.
  • the serogroup Y saccharide is similar to the serogroup W135 saccharide, except that the disaccharide repeating unit includes glucose instead of galactose.
  • the MenW135 and MenY saccharides have variable O-acetylation, but at sialic acid 7 and 9 positions [56]. Any such chemical modifications preferably take place before conjugation, but may alternatively or additionally take place during conjugation.
  • Saccharides from different serogroups are preferably purified separately, and may then be combined, either before or after conjugation.
  • Conjugates of the invention may include a serogroup A capsular saccharide antigen.
  • the saccharide can be purified and conjugated in the same way as for serogroups C, W135 and Y (see above), although it is structurally different - whereas the capsules of serogroups C, W135 and Y are based around sialic acid (N-acetyl-neuraminic acid, NeuAc), the capsule of serogroup A is based on N-acetyl-mannosamine, which is the natural precursor of sialic acid.
  • the serogroup A saccharide is particularly susceptible to hydrolysis, and its instability in aqueous media means that (a) the immunogenicity of liquid vaccines against serogroup A declines over time, and (b) quality control is more difficult, due to release of saccharide hydrolysis products into the vaccine.
  • Native MenA capsular saccharide is a homopolymer of (al ⁇ 6)-linked N-acetyl-D-mannosamine- 1 - -phosphate, with partial O-acetylation at C3 and C4.
  • the principal glycosidic bond is a 1-6 phosphodiester bond involving the hemiacetal group of CI and the alcohol group of C6 of the D-mannosamine.
  • the average chain length is 93 monomers. It has the following formula:
  • a modified saccharide antigen has been prepared which retains the immunogenic activity of the native serogroup A saccharide but which is much more stable in water. Hydroxyl groups attached at carbons 3 and 4 of the monosaccharide units are replaced by a blocking group [refs. 57 and 58].
  • the number of monosaccharide units having blocking groups in place of hydroxyls can vary. For example, all or substantially all the monosaccharide units may have blocking groups. Alternatively, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90% of the monosaccharide units may have blocking groups. At least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29 or 30 monosaccharide units may have blocking groups.
  • the number of blocking groups on a monosaccharide unit may vary.
  • the number of blocking groups on any particular monosaccharide unit may be 1 or 2.
  • the terminal monosaccharide unit may or may not have a blocking group instead of its native hydroxyl. It is preferred to retain a free anomeric hydroxyl group on a terminal monosaccharide unit in order to provide a handle for further reactions (e.g. conjugation).
  • Anomeric hydroxyl groups can be converted to amino groups (-NH 2 or -NH-E, where E is a nitrogen protecting group) by reductive amination (using, for example, NaBH 3 CN/NH 4 Cl), and can then be regenerated after other hydroxyl groups have been converted to blocking groups.
  • Blocking groups to replace hydroxyl groups may be directly accessible via a derivatizing reaction of the hydroxyl group i.e. by replacing the hydrogen atom of the hydroxyl group with another group.
  • Suitable derivatives of hydroxyl groups which act as blocking groups are, for example, carbamates, sulfonates, carbonates, esters, ethers (e.g. silyl ethers or alkyl ethers) and acetals.
  • Some specific examples of such blocking groups are allyl, alloc, benzyl, BOM, t-butyl, trityl, TBS, TBDPS, TES, TMS, TIPS, PMB, MEM, MOM, MTM, THP, etc.
  • blocking groups that are not directly accessible and which completely replace the hydroxyl group include Ci_i 2 alkyl, C 3 _i 2 alkyl, C 5 _i 2 aryl, C5-12 aryl-d_ 6 alkyl, NR*R 2 (R 1 and R 2 are defined in the following paragraph), H, F, CI, Br, C0 2 H, C0 2 (Ci_6 alkyl), CN, CF 3 , CC1 3 , etc.
  • Typical blocking groups are of the formula: -O-X-Y or -OR 3 wherein: X is C(O), S(O) or S0 2 ; Y is Ci_i 2 alkyl, Ci_i 2 alkoxy, C3_i 2 cycloalkyl, C5_i 2 aryl or C5_i 2 aryl-Ci-6 alkyl, each of which may optionally be substituted with 1 , 2 or 3 groups independently selected from F, CI, Br, C0 2 H, C0 2 (Ci_ 6 alkyl), CN, CF 3 or CC1 3 ; or Y is NR*R 2 ; R 1 and R 2 are independently selected from H, d_ 12 alkyl, C 3 _i 2 cycloalkyl, C 5 _i 2 aryl, C 5 _i 2 aryl-Ci_ 6 alkyl; or R 1 and R 2 may be joined to form a C 3 _ J2 saturated heterocyclic group; R 3 is Ci
  • R 3 is Ci_i 2 alkyl or C 3 _i 2 cycloalkyl, it is typically substituted with 1 , 2 or 3 groups as defined above.
  • R 1 and R 2 are joined to form a C 3 _i 2 saturated heterocyclic group, it is meant that R 1 and R 2 together with the nitrogen atom form a saturated heterocyclic group containing any number of carbon atoms between 3 and 12 (e.g. C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , Ci 0 , Cn, C J2 ).
  • the heterocyclic group may contain 1 or 2 heteroatoms (such as N, O or S) other than the nitrogen atom.
  • Examples of C 3 _i 2 saturated heterocyclic groups are pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, imidazolidinyl, azetidinyl and aziridinyl.
  • Blocking groups -O-X-Y and -OR 3 can be prepared from -OH groups by standard derivatizing procedures, such as reaction of the hydroxyl group with an acyl halide, alkyl halide, sulfonyl halide, etc.
  • the oxygen atom in -O-X-Y is usually the oxygen atom of the hydroxyl group
  • the -X-Y group in -O-X-Y usually replaces the hydrogen atom of the hydroxyl group.
  • the blocking groups may be accessible via a substitution reaction, such as a Mitsonobu-type substitution.
  • a substitution reaction such as a Mitsonobu-type substitution.
  • Specific blocking groups for use in the invention are -OC(0)CF 3 [59] and a carbamate group OC(0)NR 1 R 2 , where R 1 and R 2 are independently selected from Ci_6 alkyl. Typically, R 1 and R 2 are both methyl i.e. the blocking group is -OC(0)NMe 2 .
  • Carbamate blocking groups have a stabilizing effect on the glycosidic bond and may be prepared under mild conditions.
  • a particularly preferred blocking group is -OC(0)CH 3 [58].
  • the proportion of 4- and/or 3-positions in the modified N. meningitidis serogroup A saccharide that have this blocking group may vary.
  • the proportion of 4-positions that have blocking groups may be about 0%, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or about 100%, with at least 80% and about 100%) being preferred.
  • the proportion of 3-positions that have blocking groups may be about 0%, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or about 100%, with at least 80%) and about 100%) being preferred.
  • the proportion of 4- and 3-positions that have blocking groups is about the same at each position.
  • the ratio of 4-positions that have blocking groups to 3-positions that have blocking groups is about 1:1.
  • the proportion of 4-positions that have blocking groups may vary relative to the proportion of 3-positions that have blocking groups.
  • the ratio of 4-positions that have blocking groups to 3-positions that have blocking groups maybe 1:20, 1:19, 1:18, 1:17, 1:16, 1:15, 1:14, 1:13, 1:12, 1:11, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3 or 1:2.
  • the ratio of 3-positions that have blocking groups to 4-positions that have blocking groups may be 1:20, 1:19, 1:18, 1:17, 1:16, 1:15, 1:14, 1:13, 1:12, 1:11, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3 or 1:2.
  • Typical modified MenA saccharides contain n monosaccharide units, where at least h% of the monosaccharide units do not have -OH groups at both of positions 3 and 4.
  • the value of A is 24 or more ⁇ e.g.25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 98, 99 or 100) and is usually 50 or more.
  • the absent -OH groups are blocking groups as defined above.
  • modified MenA saccharides comprise monosaccharide units, wherein at least s of the monosaccharide units do not have -OH at the 3 position and do not have -OH at the 4 position.
  • the value of s is at least 1 (e.g.2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 60, 70, 80, 90).
  • the absent -OH groups are blocking groups as defined above.
  • Suitable modified MenA saccharides for use with the invention have the formula:
  • n is an integer from 1 to 100 (particularly an integer from 5 to 25, usually 15-25);
  • T is of the formul
  • each Z group is independently selected from OH or a blocking group as defined above; and each Q group is independently selected from OH or a blocking group as defined above;
  • Y is selected from OH or a blocking group as defined above;
  • E is H or a nitrogen protecting group
  • E in formula (B) is a linker or a carrier molecule of the invention. When E is a linker, the linker may be covalently bonded to a carrier molecule of the invention.
  • Each of the n+2 Z groups may be the same or different from each other.
  • each of the n+2 Q groups may be the same or different from each other.
  • All the Z groups may be OH.
  • at least 10%, 20, 30%, 40%, 50% or 60% of the Z groups may be OAc.
  • about 70% of the Z groups are OAc, with the remainder of the Z groups being OH or blocking groups as defined above.
  • At least about 7% of Q groups are blocking groups.
  • at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or even 100% of the Q groups are blocking groups.
  • the saccharide may be a glucan.
  • Glucans are glucose-containing polysaccharides found inter alia in fungal cell walls.
  • the a-glucans include one or more a-linkages between glucose subunits, whereas ⁇ -glucans include one or more ⁇ -linkages between glucose subunits.
  • the glucan used in accordance with the invention includes ⁇ linkages, and may contain only ⁇ linkages (i.e. no a linkages).
  • the glucan may comprise one or more ⁇ - 1,3 -linkages and/or one or more ⁇ -1,6-1 ⁇ 3 ⁇ 468. It may also comprise one or more ⁇ - 1 ,2-linkages and/or P-l,4-linkages, but normally its only ⁇ linkages will be ⁇ - 1,3 -linkages and /or ⁇ -1,6-1 ⁇ 3 ⁇ 468.
  • the glucan may be branched or linear.
  • soluble glucans are insoluble and have a molecular weight in the megadalton range. It is preferred to use soluble glucans in conjugates of the invention. Solubilisation may be achieved by fragmenting long insoluble glucans. This may be achieved by hydrolysis or, more conveniently, by digestion with a glucanase (e.g. with a ⁇ -l,3-glucanase or a ⁇ - ⁇ , ⁇ -glucanase). As an alternative, short glucans can be prepared synthetically by joining monosaccharide building blocks.
  • Low molecular weight glucans are preferred, particularly those with a molecular weight of less than 100 kDa (e.g. less than 80, 70, 60, 50, 40, 30, 25, 20, or 15 kDa). It is also possible to use oligosaccharides e.g. containing 60 or fewer (e.g. 59, 58, 57, 56, 55, 54, 53, 52, 51, 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4) glucose monosaccharide units. Within this range, oligosaccharides with between 10 and 50 or between 20 and 40 monosaccharide units are preferred.
  • the glucan may be a fungal glucan.
  • a 'fungal glucan' will generally be obtained from a fungus but, where a particular glucan structure is found in both fungi and non-fungi (e.g. in bacteria, lower plants or algae) then the non-fungal organism may be used as an alternative source.
  • the glucan may be derived from the cell wall of a Candida, such as C. albicans, or from Coccidioides immitis, Trichophyton verrucosum, Blastomyces dermatidis, Cryptococcus neoformans, Histoplasma capsulatum, Saccharomyces cerevisiae, Paracoccidioides brasiliensis, or Pythiumn insidiosum.
  • ⁇ -glucans there are various sources of fungal ⁇ -glucans.
  • pure ⁇ -glucans are commercially available e.g. pustulan (Calbiochem) is a ⁇ - ⁇ , ⁇ -glucan purified from Umbilicaria papullosa.
  • ⁇ -glucans can be purified from fungal cell walls in various ways.
  • Reference 60 discloses a two-step procedure for preparing a water-soluble ⁇ -glucan extract from Candida, free from cell-wall mannan, involving NaCIO oxidation and DMSO extraction.
  • the resulting product ('Candida soluble ⁇ -D-glucan' or 'CSBG') is mainly composed of a linear with a linear ⁇ - ⁇ , ⁇ -glucan moiety.
  • reference 61 discloses the production of GG-zym from C. albicans.
  • Such glucans from C. albicans include (a) ⁇ - ⁇ , ⁇ -glucans with ⁇ -l,3-glucan lateral chains and an average degree of polymerisation of about 30, and (b) with ⁇ - ⁇ , ⁇ -glucan lateral chains and an average degree of polymerisation of about 4.
  • the glucan is a ⁇ -1,3 glucan with some ⁇ -1,6 branching, as seen in e.g. laminarins. Laminarins are found in brown algae and seaweeds.
  • the ⁇ (1-3): ⁇ (1-6) ratios of laminarins vary between different sources e.g. it is as low as 3:2 in Eisenia bicyclis laminarin, but as high as 7:1 in Laminaria digititata laminarin [62].
  • the glucan used with the invention may have a ⁇ (1-3): ⁇ (1-6) ratio of between 1.5:1 and 7.5:1 e.g. about 2:1, 3:1, 4:1, 5:1, 6:1 or 7:1.
  • the glucan may have a terminal mannitol subunit, e.g. a 1,1-a-linked mannitol residue [63].
  • the glucan may also comprise mannose subunits.
  • the glucan has exclusively or mainly ⁇ -1,3 linkages, as seen in curdlan. These glucans may elicit better protection than glucans comprising other linkages, particularly glucans comprising ⁇ -1,3 linkages and a greater proportion of ⁇ -1,6 linkages.
  • the glucan may be made solely of -l,3-linked glucose residues (e.g. linear ⁇ -D-glucopyranoses with exclusively 1,3 linkages).
  • the glucan may include monosaccharide residues that are not ⁇ - 1,3-linked glucose residues e.g. it may include -l,6-linked glucose residues.
  • the ratio of ⁇ - 1,3-linked glucose residues to these other residues should be at least 8:1 (e.g. >9:1, > 10: 1 , >11:1, >12:1, >13:1, >14:1, >15:1, >16:1, >17:1, >18:1, >19:1, >20:1, >25:1, >30:1, >35:1, >40:1, >45:1, >50: 1, >75: 1, >100: 1, etc.) and/or there are one or more (e.g. >1, >2, >3, >4, >5, >6, >7, >8, >9, >10, >11, >12, etc.) sequences of at least five (e.g.
  • non-terminal residues adjacent non-terminal residues linked to other residues only by ⁇ -1,3 linkages.
  • non-terminal it is meant that the residue is not present at a free end of the glucan.
  • the adjacent non-terminal residues may not include any residues coupled to a carrier molecule, linker or other spacer as described below. The presence of five adjacent non-terminal residues linked to other residues only by ⁇ -1,3 linkages may provide a protective antibody response, e.g. against C. albicans.
  • a conjugate may include two different glucans e.g. a first glucan having a ⁇ (1-3): ⁇ (1-6) ratio of between 1.5:1 and 7.5:1, and a second glucan having exclusively or mainly ⁇ -1,3 linkages.
  • a conjugate may include both a laminarin glucan and a curdlan glucan.
  • a ⁇ -glucan includes both ⁇ -1,3 and ⁇ -1,6 linkages at a desired ratio and/or sequence
  • this glucan may be found in nature (e.g. a laminarin), or it may be made artificially. For instance, it may be made by chemical synthesis, in whole or in part. Methods for the chemical synthesis of ⁇ -1,3/ ⁇ -1,6 glucans are known, for example from references 64-74.
  • ⁇ -glucan including both ⁇ -1,3 and ⁇ - 1 ,6 linkages at a desired ratio may also be made starting from an available glucan and treating it with a ⁇ -l,6-glucanase (also known as glucan endo-l,6 ⁇ -glucosidase, l,6 ⁇ -D-glucan glucanohydrolase, etc.; EC 3.2.1.75) or a ⁇ -1,3 ⁇ 3 ⁇ 4 ⁇ 8 ⁇ (such as an exo-l,3-glucanase (EC 3.2.1.58) or an endo-l,3-glucanase (EC 3.2.1.39) until a desired ratio and/or sequence is reached.
  • a ⁇ -l,6-glucanase also known as glucan endo-l,6 ⁇ -glucosidase, l,6 ⁇ -D-glucan glucanohydrolase, etc.
  • EC 3.2.1.75 also known as glucan endo-l,
  • ⁇ - 1 ,6-glucanase treatment may be pursued to completion, as ⁇ -1,6 ⁇ 3 ⁇ 4 ⁇ 8 ⁇ will eventually yield pure ⁇ -1,3 glucan.
  • a pure P-l,3-glucan may be used. These may be made synthetically, by chemical and/or enzymatic synthesis e.g. using a (l ⁇ 3)- -D-glucan synthase, of which several are known from many organisms (including bacteria, yeasts, plants and fungi). Methods for the chemical synthesis of ⁇ - 1,3 glucans are known, for example from references 75-78.
  • a natural -l,3-glucan may be used, such as a curdlan (linear -l,3-glucan from an Agrobacterium previously known as Alcaligenes faecalis var. myxogenes; commercially available e.g. from Sigma-Aldrich catalog C7821) or paramylon ( -l,3-glucan from Euglend).
  • Curdlan linear -l,3-glucan from an Agrobacterium previously known as Alcaligenes faecalis var. myxogenes; commercially available e.g. from Sigma-Aldrich catalog C7821
  • paramylon -l,3-glucan from Euglend.
  • Organisms producing high levels of ⁇ - 1,3 -glucans are known in the art e.g. the Agrobacterium of refs. 79 & 80, or the Euglena gracilis of ref. 81.
  • Laminarin and curdlan are typically found in nature as high molecular weight polymers e.g. with a molecular weight of at least lOOkDa. They are often insoluble in aqueous media. In their natural forms, therefore, they are not well suited to immunisation.
  • the invention may use a shorter glucan e.g. those containing 60 or fewer glucose monosaccharide units (e.g.
  • a glucan having a number of glucose residues in the range of 2-60 may be used e.g. between 10-50 or between 20-40 glucose units.
  • a glucan with 25-30 glucose residues is particularly useful.
  • Suitable glucans may be formed e.g. by acid hydrolysis of a natural glucan, or by enzymatic digestion e.g.
  • glucanase such as a ⁇ - 1,3 -glucanase.
  • a glucan with 11-19, e.g. 13-19 and particularly 15 or 17, glucose monosaccharide units is also useful.
  • glucans with the following structures (A) or (B) are specifically envisaged for use in the present invention:
  • n+2 is in the range of 2-60, e.g. between 10-50 or between 2-40.
  • n+2 in the range of 25-30 or 11-19, e.g. 13-17.
  • n is in the range of 0-9, e.g. between 1-7 or between 2-6.
  • n is in the range of 3-4 or 1-3.
  • the glucan is a single molecular species.
  • all of the glucan molecules are identical in terms of sequence. Accordingly, all of the glucan molecules are identical in terms of their structural properties, including molecular weight etc.
  • this form of glucan is obtained by chemical synthesis, e.g. using the methods described above.
  • reference 76 describes the synthesis of a single ⁇ -1,3 linked species.
  • the glucan may be obtained from a natural glucan, e.g. a glucan from Laminaria digitata, Agrobacterium or Euglena as described above, with the glucan being purified until the required single molecular species is obtained.
  • Natural glucans that have been purified in this way are commercially available.
  • a glucan that is a single molecular species may be identified by measuring the polydispersity (Mw/Mn) of the glucan sample. This parameter can conveniently be measured by SEC-MALLS, for example as described in reference 82.
  • Suitable glucans for use in this embodiment of the invention have a polydispersity of about 1, e.g. 1.01 or less.
  • Solubility of natural glucans, such as curdlan can be increased by introducing ionic groups (e.g. by sulfation, particularly at 0-6 in curdlan). Such modifications may be used with the invention, but are ideally avoided as they may alter the glucan's antigenicity.
  • the saccharide is a glucan, it is typically a laminarin.
  • the saccharide may also be a bacterial capsular saccharide.
  • Further exemplary bacterial capsular saccharides include those from Streptococcus pneumoniae.
  • the saccharide is a capsular saccharides from S. pneumoniae
  • it is typically from one of the following pneumococcal serotypes: 1, 2, 3, 4, 5, 6A, 6B, 7F, 8, 9N, 9V, 10A, 11A, 12F, 14, 15B, 17F, 18C, 19A, 19F, 20, 22F, 23F and 33F, preferably from 1, 5, 6B, 14, 19F and 23F.
  • Capsular polysaccharides from S. pneumoniae comprise repeating oligosaccharide units which may contain up to 8 sugar residues. The oligosaccharide units for the main S. pneumoniae serotypes are described in and refs 83 and 84.
  • exemplary bacterial capsular saccharides include those from Staphylococcus aureus, particularly the capsular polysaccharides of S. aureus type 5 and type 8.
  • the structures of type 5 and type 8 capsular polysaccharides were described in references 85 and 86 as:
  • Type 5 ⁇ 4)-p-D-ManNAcA(30Ac)-(l ⁇ 4)-a-L-FucNAc(l ⁇ 3)- -D-FucNAc-(l ⁇
  • Type 8 ⁇ 3)-p-D-ManNAcA(40Ac)-(l ⁇ 3)-a-L-FucNAc(l ⁇ 3)- -D-FucNAc-(l ⁇
  • Type 5 ⁇ 4)- -D-ManNAcA-(l ⁇ 4)-a-L-FucNAc(30Ac)-(l ⁇ 3)- -D-FucNAc-(l ⁇
  • Type 8 ⁇ 3)-p-D-ManNAcA(40Ac)-(l ⁇ 3)-a-L-FucNAc(l ⁇ 3)-a-D-FucNAc(l ⁇ ).
  • the polysaccharide may be chemically modified relative to the capsular polysaccharide as found in nature.
  • the polysaccharide may be de-O-acetylated (partially or fully), de-N-acetylated (partially or fully), N-propionated (partially or fully), etc.
  • De-acetylation may occur before, during or after conjugation, but typically occurs before conjugation.
  • de-acetylation may or may not affect immunogenicity e.g. the NeisVac-CTM vaccine uses a de-O-acetylated polysaccharide, whereas MenjugateTM is acetylated, but both vaccines are effective.
  • the effect of de-acetylation etc. can be assessed by routine assays. For example, the relevance of O-acetylation on S.
  • aureus type 5 or type 8 capsular polysaccharides is discussed in reference 88.
  • the native polysaccharides are said in this document to have 75% O-acetylation.
  • These polysaccharides induced antibodies to both the polysaccharide backbone and O-acetyl groups.
  • Polysaccharides with 0% Oacetylation still elicited antibodies to the polysaccharide backbone. Both types of antibody were opsonic against S. aureus strains that varied in their O-acetyl content.
  • the type 5 or type 8 capsular polysaccharides used in the present invention may have between 0 and 100% O-acetylation.
  • the degree of Oacetylation of the polysaccharide can be determined by any method known in the art, for example, by proton NMR (e.g. as described in references 89, 90, 91 or 92). A further method is described in reference 93. Similar methods may be used to determine the degree of N-acetylation of the polysaccharide.
  • O-acetyl groups may be removed by hydrolysis, for example by treatment with a base such as anhydrous hydrazine [94] or NaOH [88]. Similar methods may be used to remove N-acetyl groups.
  • treatments that lead to hydrolysis of the O-acetyl groups are minimised, e.g. treatments at extremes of pH.
  • Capsular polysaccharides can be purified by known techniques, as described in the references herein.
  • a typical process involves phenol-ethanol inactivation of S. aureus cells, centrifugation, lysostaphin treatment, RNase/DNase treatment, centrifugation, dialysis, protease treatment, further dialysis, filtration, precipitation with ethanol/CaCl 2 , dialysis, freeze-drying, anion exchange chromatography, dialysis, freeze-drying, size exclusion chromatography, dialysis and freeze-drying [95].
  • An alternative process involves autoclaving S.
  • aureus cells ultrafiltration of the polysaccharide- containing supernatant, concentration, lyophilisation, treatment with sodium metaperiodate to remove teichoic acid, further ultrafiltration, diafiltration, high performance size exclusion liquid chromatography, dialysis and freeze-drying [96].
  • the invention is not limited to polysaccharides purified from natural sources, however, and the polysaccharides may be obtained by other methods, such as total or partial synthesis.
  • Other bacterial capsular saccharides such as Bacillus subtilis
  • exemplary bacterial capsular saccharides include those from Haemophilus influenzae Type b, Salmonella enterica Typhi Vi and Clostridium difficile.
  • the invention may also use non-capsular bacterial saccharides.
  • An exemplary non-capsular bacterial saccharide is the Streptococcus pyogenes GAS carbohydrate (also known as the GAS cell wall polysaccharide, or GASP).
  • GAS carbohydrate also known as the GAS cell wall polysaccharide, or GASP.
  • This saccharide features a branched structure with an L-rhamnopyranose (Rha/?) backbone consisting of alternating alpha-(l ⁇ 2) and alpha-(l ⁇ 3) links and D-N- acetylglucosamine (Glc/MAc) residues beta-(l ⁇ 3)-connected to alternating rhamnose rings ( [97]).
  • the GAS carbohydrate will generally be in its native form, but it may have been modified.
  • the saccharide may be shorter than the native GAS carbohydrate, or may be chemically modified.
  • the saccharide used according to the invention may be a substantially full-length GAS carbohydrate, as found in nature, or it may be shorter than the natural length.
  • Full-length polysaccharides may be depolymerised to give shorter fragments for use with the invention e.g. by hydrolysis in mild acid, by heating, by sizing chromatography, etc.
  • a short fragment thought to correspond to the terminal unit on the GAS carbohydrate has been proposed for use in a vaccine [98]. Accordingly, short fragments are envisaged in the present invention.
  • the GAS carbohydrate typically has a molecular weight of about 10, in particular about 7.5-8.5 kDa.
  • Molecular masses can be measured by HPLC, for example SEC-HPLC using a TSK Gel G3000SW column (Sigma) relative to pullulan standards, such as those available from Polymer Standard Service [99] .
  • the saccharide may be chemically modified relative to the GAS carbohydrate as found in nature.
  • the saccharide may be de-N-acetylated (partially or fully), N-propionated (partially or fully), etc.
  • the effect of de-acetylation etc., for example on immunogenicity can be assessed by routine assays.
  • Capsular saccharides may be purified by known techniques, for example as described in Wessels et al. (J Clin Invest, 1990 86: 1428-33) or Wessels et al. (Infect Immun 1989, 57: 1089-94).
  • a typical purification process involves base extraction, centrifugation, filtration, RNase/DNase treatment, protease treatment, concentration, size exclusion chromatography, ultrafiltration, anion exchange chromatography, and further ultrafiltration.
  • Treatment of GBS cells with the enzyme mutanolysin which cleaves the bacterial cell wall to free the cell wall components, is also useful.
  • the purification process described in WO2006/082527 can be used. This involves base extraction, ethanol/CaCl 2 treatment, CTAB precipitation, and re solubilisation. Further extraction and purification processes are known in the art, such as those described in WO 2009/081276.
  • the invention involves a conjugate comprising an antigen and a carrier molecule, wherein the carrier molecule comprises a spbl polypeptide and at least one BP-2a polypeptide.
  • the carrier molecule is a fusion protein which comprises an spbl polypeptide and at least one BP-2a polypeptide, at least two BP-2a polypeptides, particularly three, four, five, six or seven BP-2a polypeptides or fragments thereof.
  • the carrier molecule may be covalently conjugated to the antigen directly or via a linker. Any suitable conjugation reaction can be used, with any suitable linker where desired.
  • Attachment of the antigen to the carrier is preferably via a -NH group e.g. in the side chain of a lysine residue in a carrier protein, or of an arginine residue. Where the antigen has a free aldehyde group, then this can react with an amine in the carrier to form a conjugate by reductive amination. Attachment to the carrier may also be via a -SH group e.g. in the side chain of a cysteine residue. Alternatively the antigen may be attached to the carrier via a linker molecule.
  • the antigen will typically be activated or functionalised prior to conjugation. Activation may involve, for example, cyanylating reagents such as CDAP ⁇ e.g. 1 -cyano-4-dimethylamino pyridinium tetrafluoroborate [100, 101 , etc.]).
  • cyanylating reagents such as CDAP ⁇ e.g. 1 -cyano-4-dimethylamino pyridinium tetrafluoroborate [100, 101 , etc.]).
  • Other suitable techniques use carbodiimides, hydrazides, active esters, norborane, p-nitrobenzoic acid, N-hydroxysuccinimide, S-NHS, EDC, TSTU (see also the introduction to reference 7).
  • Direct linkages to the protein may comprise oxidation of the antigen followed by reductive amination with the protein, as described in, for example, refs. 102 and 103.
  • Linkages via a linker group may be made using any known procedure, for example, the procedures described in refs. 104 and 105.
  • the linker is attached via the anomeric carbon of a saccharide antigen.
  • a preferred type of linkage is an adipic acid linker, which may be formed by coupling a free -NH 2 group (e.g. introduced to a saccharide by amination) with adipic acid (using, for example, diimide activation), and then coupling a protein to the resulting antigen-adipic acid intermediate [5, 106, 107].
  • a similar preferred type of linkage is a glutaric acid linker, which may be formed by coupling a free -NH 2 group with glutaric acid in the same way.
  • Adipid and glutaric acid linkers may also be formed by direct coupling to the antigen, i. e. without prior introduction of a free group, e.g. a free -NH 2 group, to the antigen, followed by coupling a protein to the resulting antigen- adipic/glutaric acid intermediate.
  • a free group e.g. a free -NH 2 group
  • Another preferred type of linkage is a carbonyl linker, which may be formed by reaction of a free hydroxyl group of a modified antigen with CDI [108, 109] followed by reaction with a protein to form a carbamate linkage.
  • linkers include ⁇ -propionamido [1 10], nitrophenyl-ethylamine [1 1 1], haloacyl halides [1 12], glycosidic linkages [1 13], 6-aminocaproic acid [1 14], N-succinimidyl-3-(2-pyridyldithio)-propionate (SPDP) [1 15], adipic acid dihydrazide ADH [1 16], C 4 to Ci 2 moieties [1 17], etc. Carbodiimide condensation can also be used [1 18].
  • a bifunctional linker may be used to provide a first group for coupling to an amine group in the antigen (e.g.
  • the first group is capable of direct coupling to the antigen, i.e. without prior introduction of a group, e.g. an amine group, to the antigen.
  • the first group in the bifunctional linker is thus able to react with an amine group (-NH 2 ) on the antigen. This reaction will typically involve an electrophilic substitution of the amine's hydrogen.
  • the first group in the bifunctional linker is able to react directly with the antigen.
  • the second group in the bifunctional linker is typically able to react with an amine group on the carrier. This reaction will again typically involve an electrophilic substitution of the amine.
  • Preferred L 2 groups are straight chain alkyls with 1 to 10 carbon atoms (e.g. Ci, C 2 , C3, C 4 , C5, C , C , C , C9, Cio) e.g. -(CH ) 4 - or -(CH ) 3 -.
  • reaction with the antigen involves direct coupling and the reaction with the carrier involves an amine then it is also preferred to use a bifunctional linker.
  • a homobifunctional linker of the formula X-L-X may be used, where: the two X groups are the same as each other and can react with the antigen/amine; and where L is a linking moiety in the linker.
  • a heterobifunctional linker of the formula X-L-X may be used, where: the two X groups are different and one can react with the antigen while the other can react with the amine; and where L is a linking moiety in the linker.
  • a preferred X group is N-oxysuccinimide.
  • L preferably has formula
  • L'-L 2 -L' where L' is carbonyl.
  • Preferred L 2 groups are straight chain alkyls with 1 to 10 carbon atoms
  • Ci e.g. Ci, C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , Cio
  • Cio e.g. -(CH 2 ) 4 - or -(CH 2 ) 3 -.
  • X groups for use in the bifunctional linkers described in the two preceding paragraphs are those which form esters when combined with HO-L-OH, such as norborane, p-nitrobenzoic acid, and sulfo-N-hydroxysuccinimide.
  • Further bifunctional linkers for use with the invention include acryloyl halides (e.g. chloride) and haloacylhalides.
  • the linker will generally be added in molar excess to antigen during coupling to the antigen.
  • the resultant conjugate may form a "star" structure.
  • This structure comprises a central carrier molecule with multiple antigen molecules radiating from the carrier (optionally via linkers).
  • the antigen has more than one group that is linked to the carrier molecule (optionally via a linker)
  • the carrier has more than one group that is linked to different antigen/linker molecules
  • the resultant conjugate may form a "net” structure.
  • This structure comprises a network of carrier molecules connected by antigen molecules (optionally via linkers).
  • Conjugates may have excess carrier (w/w) or excess antigen (w/w) e.g. in the ratio range of 1 :5 to 5: 1. Conjugates with excess carrier protein are typical e.g. in the range 0.2: 1 to 0.9: 1, or equal weights.
  • the conjugate may include small amounts of free (i.e. unconjugated) carrier. When a given carrier protein is present in both free and conjugated form in a composition of the invention, the unconjugated form is preferably no more than 5% of the total amount of the carrier protein in the composition as a whole, and more preferably present at less than 2% (by weight).
  • the composition may also comprise free carrier protein as immunogen [119].
  • free and conjugated antigens can be separated.
  • suitable methods e.g. hydrophobic chromatography, tangential ultrafiltration, diafiltration, etc. [see also refs. 120, 121 etc. ] . Tangential flow ultrafiltration is preferred.
  • a saccharide moiety in the conjugate is preferably a low molecular weight saccharide or an oligosaccharide, as defined above. Oligosaccharides will typically be sized prior to conjugation.
  • the conjugate is preferably soluble in water and/or in a physiological buffer. Conjugation
  • the invention involves conjugates that are capsular saccharides from GBS serotypes la, lb, II, III and V, each conjugated to a carrier protein.
  • covalent conjugation of saccharides to carriers enhances the immunogenicity of saccharides as it converts them from T-independent antigens to T-dependent antigens, thus allowing priming for immunological memory.
  • Conjugation is particularly useful for paediatric vaccines and is a well-known technique. Conjugation of GBS saccharides has been widely reported e.g. see Paoletti et al. (1990) J Biol Chem 265: 18278-83.
  • polysaccharides are immunogenic
  • conjugation of polysaccharides to carrier proteins can improve or enhance immunogenicity.
  • carrier refers to an immunogenic substance which, when conjugated to an antigen (such as a polysaccharide) and administered to a suitable animal, will induce or enhance an immune response in the animal, particularly a protective immune response, and elicit the production of antibodies that bind specifically to the antigen, for example, the above described polysaccharides.
  • an antigen such as a polysaccharide
  • the typical prior art process for GBS saccharide conjugation typically involves reductive amination of a purified saccharide to a carrier protein such as tetanus toxoid (TT) or CRM197 Wessels et al. (1990) J Clin Invest 86: 1428-33.
  • the reductive amination involves an amine group on the side chain of an amino acid in the carrier and an aldehyde group in the saccharide.
  • GBS capsular saccharides do not include an aldehyde group in their natural form then this is typically generated before conjugation by oxidation (e.g. periodate oxidation) of a portion (e.g. between 5 and 40%, particularly between 10 and 30%, preferably about 20%)) of the saccharide's sialic acid residues.
  • Conjugate vaccines prepared in this manner have been shown to be safe and immunogenic in humans for each of GBS serotypes la, lb, II, III, and V, Paoletti & Kasper (2003) Expert Opin Biol Ther 3:975-84.
  • all of the conjugates in the immunogenic compositions of the present invention may be prepared in this manner.
  • an aldehyde group may be generated in this saccharide before conjugation by oxidation (e.g. periodate oxidation) of a portion (e.g. between 5 and 40%>, particularly between 10 and 30%>, preferably about 20%>) of the saccharide's galactose residues.
  • oxidation e.g. periodate oxidation
  • a portion e.g. between 5 and 40%>, particularly between 10 and 30%>, preferably about 20%>
  • An alternative conjugation process involves the use of -NH 2 groups in the saccharide (either from de-N-acetylation, or after introduction of amines) in conjunction with bifunctional linkers.
  • one or more of the conjugates in the immunogenic compositions of the present invention may be prepared in this manner.
  • a further alternative process utilises free aldehyde groups of terminal 2,5-anhydro-D-mannose residues from depolymerization of type II or type III capsular saccharides by mild deaminative cleavage are used for conjugation by reductive amination.
  • one or more of the conjugates in the immunogenic compositions of the present invention may be prepared in this manner.
  • the carrier and antigen may be conjugated by sortase mediated ligation, for example, as described in WO13065009 or WO13003555.
  • Conjugates of the invention may comprise utilise protein-protein conjugation and may comprise proteins conjugated to proteins.
  • the BP-2a polypeptide may be conjugated to the spbl polypeptide.
  • the protein carrier molecule of the invention may be conjugated to a protein antigen. Any appropriate method may be used to generate a conjugate of the invention. For example, suitable methods are detailed below.
  • Protein-protein linkages in the conjugates of the invention may be generated using any appropriate technique, and the skilled person will be aware of such suitable techniques.
  • suitable cross-linking agents include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA), 5,5'-dithiobis(2-nitrobenzoic acid) (DTNB), o-phenylenedimaleimide (oPDM), N-succinimidyl-3-(2- pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl)cyclohaxane- 1 - carboxylate (sulfo-SMCC) (see e.g. [122,123]).
  • Other methods include those described in refernces 124, 125 and 126.
  • Preferred conjugating agents are SATA and sulfo-SMCC, both available from Pierce Chemical Co. (Rockford, IL
  • Peptide-protein carrier conjugates may also be formed using conventional cross-linking agents such as carbodimides.
  • carbodimides are 1 -cyclohexyl-3-(2-morpholinyl-(4-ethyl) carbodiimide (CMC), 1 -ethyl-3-(3-dimethyaminopropyl) carbodiimide (EDC) and 1 -ethyl-3-(4- azonia-44-dimethylpentyl) carbodiimide.
  • CMC cyclohexyl-3-(2-morpholinyl-(4-ethyl) carbodiimide
  • EDC 1 -ethyl-3-(3-dimethyaminopropyl) carbodiimide
  • EDC 1 -ethyl-3-(4- azonia-44-dimethylpentyl) carbodiimide.
  • other suitable cross-linking agents are cyanogen bromide, glut
  • any of a number of homo- bifunctional agents including a homo-bifunctional aldehyde, a homo-bifunctional epoxide, a homo- bifunctional imido-ester, a homo-bifunctional N-hydroxysuccinimide ester, a homo-bifunctional maleimide, a homo-bifunctional alkyl halide, a homo-bifunctional pyridyl disulfide, a homo- bifunctional aryl halide, a homo-bifunctional hydrazide, a homo-bifunctional diazonium derivative and a homo-bifunctional photoreactive compound may be used.
  • hetero- bifunctional compounds for example, compounds having an amine-reactive and a sulfhydryl- reactive group, compounds with an amine-reactive and a photoreactive group and compounds with a carbonyl-reactive and a sulfhydryl-reactive group.
  • homo-bifunctional cross-linking agents include the bifunctional N- hydroxysuccinimide esters dithiobis(succinimidylpropionate), disuccinimidyl suberate, and disuccinimidyl tartrate; the bifunctional imido-esters dimethyl adipimidate, dimethyl pimelimidate, and dimethyl suberimidate; the bifunctional sulfhydryl-reactive crosslinkers l ,4-di-[3'-(2'- pyridyldithio)propionamido]butane, bismaleimidohexane, and bis-N-maleimido-l ,8-octane; the bifunctional aryl halides l ,5-difluoro-2,4-dinitrobenzene and 4,4'-difluoro-3,3'-dinitrophenylsulfone; bifunctional photoreactive agents such as ?zs-[P
  • SMCC succinimidyl-4-(N- maleimidomethyl)cyclohexane- 1 -carboxylate
  • MBS m-maleimidobenzoyl-N-hydroxysuccinimide ester
  • SIAB N-succinimidyl(4-iodoacteyl)aminobenzoate
  • SMPB succinimidyl-4-(p- maleimidophenyl)butyrate
  • GMBS N-(y-maleimidobutyryloxy)succinimide ester
  • MPBH 4-(4-N- maleimidopohenyl)butyric acid hydrazide
  • M2C2H 4-(N-maleimidomethyl)cyclohexane-l - carboxyl-hydrazide
  • SMPT succin
  • non-natural amino acid residues are to be incorporated into the carrier molecule.
  • this can be performed using standard procedures.
  • One such method comprises the use of modified host cells in which the amino acyl tRNA synthetase for a specific codon has been engineered to conjugate the tRNA to a non-natural amino acid which is then incorporated into the carrier during translation [see ref. 127 for a review of such techniques].
  • some procedures exploit the fact that some non-natural amino acids are incorporated into proteins by the native cellular machinery when the natural cognate amino acid is not present.
  • An example of this second type of procedure is observed in the incorporation of HAG. Where the non-natural amino acid is HAG, then thiyl-ene conjugation is used [see, e.g., ref. 128].
  • the conjugates of the invention may be mixed with further antigens. These further antigens may be other conjugates of the invention or they may be other antigens.
  • conjugates are envisaged.
  • the mixtures of conjugates will be provided as pharmaceutical compositions.
  • At least one of the conjugates in these mixtures is a conjugate of the invention, i.e. the carrier molecule comprises a BP-2a polypeptide and a spbl polypeptide.
  • the other conjugate(s) in these mixtures may also be conjugates of the invention.
  • the carrier molecule may be any suitable carrier protein (as described below).
  • the conjugates of the invention are mixed with conjugates comprising carrier molecules that do not comprise BP-2a polypeptides and spbl polypeptides.
  • conjugates of the invention are mixed with conjugates comprising CRMi 97 as the carrier molecule.
  • carrier molecules comprising a BP-2a polypeptide and a spbl polypeptide can be particularly effective when combined with further conjugates comprising CRMi 97 .
  • conjugates of the invention may be mixed with conjugates comprising GBS80 as a carrier molecule.
  • conjugates of the invention are mixed are mixed with conjugates comprising GBS80 and conjugates comprising CRMi 97 .
  • the other conjugates preferably comprise antigens from the same pathogen as the antigen in the conjugate of the invention.
  • mixtures are envisaged comprising a first antigen, such as a saccharide antigen, from a pathogen conjugated to a carrier comprising a BP-2a polypeptide and a spbl polypeptide, and a second antigen from the same pathogen conjugated to another carrier molecule (and possibly a third antigen from the same pathogen, and a fourth, etc.).
  • the second antigen may be derived from a different serotype. In this way it may be possible to induce broad immune response specific for numerous different antigens, making use of the advantageous properties of the carrier molecule of the invention.
  • the invention provides a pharmaceutical composition comprising a mixture of conjugates, wherein at least one conjugate comprises a carrier comprising a
  • the mixture comprises more than one capsular saccharide derived from Streptococcus agalactiae serogroups la, lb, II, III, V.
  • the mixture comprises 4, or all 5 of these saccharides.
  • it is the serotype-V saccharide, or even more particularly, the serotype-II saccharide that is conjugated to the BP-2a and spbl carrier of the invention.
  • the invention provides an immunogenic composition comprising: (a) a conjugate that is a capsular saccharide from GBS serotype la conjugated to a first carrier protein; (b) a conjugate that is a capsular saccharide from GBS serotype lb conjugated to a second carrier protein; (c) a conjugate that is a capsular saccharide from GBS serotype III conjugated to a third carrier protein; (d) a conjugate that is a capsular saccharide from GBS serotype II conjugated to a fourth carrier protein; and (e) a conjugate that is a capsular saccharide from GBS serotype V conjugated to a fifth carrier protein, wherein at least one of the first, second, third, fourth or fifth carrier proteins is a BP-2a and spbl carrier of the invention.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising S. agalactiae saccharides la, lb and III conjugated to CRM197, saccharide from serotype V S. agalactiae conjugated to GBS80, and saccharide from serotype II S. agalactiae conjugated to the BP-2a and spbl carrier of the invention.
  • the serotype II saccharide may be conjugated to GBS80
  • the serotype V saccharide may be conjugated to the BP-2a and spbl carrier of the invention.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising: (a) a conjugate that is a capsular saccharide from GBS serotype la conjugated to a first carrier protein, (b) a conjugate that is a capsular saccharide from GBS serotype lb conjugated to a second carrier protein, (c) a conjugate that is a capsular saccharide from GBS serotype III conjugated to a third carrier protein, wherein the first, second and third carrier proteins are selected from the group consisting of tetanus toxoid (TT), diphtheria toxoid (DT), GBS80, GBS67 and CRM 197 ; and, (d) a conjugate that is a capsular saccharide from GBS serotype II conjugated to a fourth carrier protein, and (e) a conjugate that is a capsular saccharide from GBS serotype V conjugated to a fifth carrier protein, wherein one of the fourth or fifth carrier proteins is selected from the group consist
  • mixtures of conjugates from more than one serogroup of Neisseria meningitidis are envisaged e.g. compositions comprising saccharides from serogroups A+C, A+W135, A+Y, C+W135, C+Y, W135+Y, A+C+W135, A+C+Y, C+W135+Y, A+C+W135+Y, etc.
  • the mixture is a mixture of conjugates comprising saccharides from serogroups A, C, W135 and Y.
  • At least one of the conjugates in these mixtures is a conjugate of the invention, i.e.
  • the carrier molecule comprises a BP-2a polypeptide and a spbl polypeptide.
  • the other conjugate(s) in these mixtures will also be conjugates of the invention.
  • the carrier molecule may be any suitable carrier protein (as described below), typically the same carrier molecule in each conjugate.
  • Suitable carrier proteins are bacterial toxins, such as diphtheria or tetanus toxins, or toxoids or mutants thereof.
  • the CRM197 diphtheria toxin mutant [129] may be suitable.
  • suitable carrier proteins include the N. meningitidis outer membrane protein complex [130], synthetic peptides [131,132], heat shock proteins [133,134], pertussis proteins [135,136], cytokines [137], lymphokines [137], hormones [137], growth factors [137], human serum albumin (typically recombinant), artificial proteins comprising multiple human CD4 + T cell epitopes from various pathogen-derived antigens [20] such as N19 [138], protein D from Haemophilus influenzae [139-141], pneumococcal surface protein PspA [142], pneumolysin [143] or its non-toxic derivatives [144], iron-uptake proteins [145], toxin A or B from Clostridium difficile [146], a GBS protein [147],
  • a single carrier protein might carry more than one polysaccharide antigen [149,150].
  • different saccharides can be mixed prior to the conjugation process.
  • the separate conjugates may be based on the same carrier, particularly the same carrier comprising a BP-2a polypeptide and a spbl polypeptide.
  • the mixtures may also comprise proteins.
  • the mixtures may comprise 5. agalactiae or proteins [e.g. 147, 151-153]
  • the further antigen(s) may comprise antigens from non-5 * , agalactiae pathogens.
  • the compositions of the invention may further comprise one or more non-5 * , agalactiae antigens, including additional bacterial, viral or parasitic antigens. These may be selected from the following: - a saccharide antigen from Streptococcus pneumoniae [e.g. refs. 154-156; chapters 22 & 23 of ref. 163].
  • an antigen from hepatitis A virus such as inactivated virus [e.g. 157, 158; chapter 15 of ref.
  • an antigen from hepatitis B virus such as the surface and/or core antigens [e.g. 158, 159; chapter 16 of ref. 163].
  • hepatitis C virus an antigen from hepatitis C virus [e.g. 160].
  • Bordetella pertussis such as pertussis holotoxin (PT) and filamentous haemagglutinin (FHA) from B. pertussis, optionally also in combination with pertactin and/or agglutinogens 2 and 3 [e.g. refs. 161 & 162; chapter 21 of ref. 163].
  • diphtheria antigen such as a diphtheria toxoid [e.g. chapter 13 of ref. 163].
  • tetanus antigen such as a tetanus toxoid [e.g. chapter 27 of ref. 163].
  • - a saccharide antigen from H. influenzae B [e.g. chapter 14 of ref. 163].
  • - a protein antigen from serogroup B ofN. meningitidis [e.g. refs. 164 to 169].
  • Neisseria gonorrhoeae an antigen from Neisseria gonorrhoeae [e.g. 164-167].
  • Chlamydia pneumoniae an antigen from Chlamydia pneumoniae [e.g. 170, 171, 172, 173, 174, 175, 176].
  • rabies antigen(s) e.g. 181
  • lyophilised inactivated virus e.g.182, RabAvertTM
  • influenza antigen(s) e.g. chapters 17 & 18 of ref. 163
  • haemagglutinin and/or neuraminidase surface proteins such as the haemagglutinin and/or neuraminidase surface proteins.
  • Streptococcus pyogenes group A streptococcus [e.g. 184, 185, 186].
  • an antigen from Staphylococcus epidermidis e.g. type I, II and/or III capsular polysaccharide obtainable from strains ATCC-31432, SE-360 and SE-10 as described in refs. 187, 188 and 189].
  • the carrier molecule may be a carrier of the invention, i. e. a carrier that comprises a BP-2a polypeptide and a spbl polypeptide.
  • the carrier molecule may be any suitable carrier protein, e.g. as described above. Conjugation of Haemophilus influenzae B, meningococcal and pneumococcal saccharide antigens is well known.
  • Toxic protein antigens may be detoxified where necessary (e.g. detoxification of pertussis toxin by chemical and/or genetic means [162]).
  • diphtheria antigen is included in the composition it is typical also to include tetanus antigen and pertussis antigens. Similarly, where a tetanus antigen is included it is typical also to include diphtheria and pertussis antigens. Similarly, where a pertussis antigen is included it is typical also to include diphtheria and tetanus antigens.
  • Antigens may be adsorbed to an aluminium salt. Antigens in the composition will typically be present at a concentration of at least ⁇ g/ml each. In general, the concentration of any given antigen will be sufficient to elicit an immune response against that antigen.
  • nucleic acid encoding the antigen may be used [e.g. refs. 190 to 198]. Protein components of the compositions of the invention may thus be replaced by nucleic acid (usually DNA e.g. in the form of a plasmid) that encodes the protein.
  • compositions of the invention there may be an upper limit to the number of antigens included in compositions of the invention.
  • the number of antigens (including conjugates of the invention) in a composition of the invention may be less than 20, less than 19, less than 18, less than 17, less than 16, less than 15, less than 14, less than 13, less than 12, less than 1 1, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, or less than 3.
  • the number of conjugates of the invention in a composition may be less than 6, less than 5, or less than 4.
  • compositions comprising the conjugates
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising (a) a conjugate of the invention, and (b) a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier is available in reference 199.
  • compositions of the invention may be prepared in various forms.
  • the compositions may be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the composition may be prepared for topical administration e.g. as an ointment, cream or powder.
  • the composition be prepared for oral administration e.g. as a tablet or capsule, or as a syrup (optionally flavoured).
  • the composition may be prepared for pulmonary administration e.g. as an inhaler, using a fine powder or a spray.
  • the composition may be prepared as a suppository or pessary.
  • the composition may be prepared for nasal, aural or ocular administration e.g. as drops, as a spray, or as a powder [e.g. 200].
  • the composition may be included in a mouthwash.
  • the composition may be lyophilised.
  • the pharmaceutical composition is preferably sterile. It is preferably pyrogen-free. It is preferably buffered e.g. at between pH 6 and pH 8, generally around pH 7.
  • the invention also provides a delivery device containing a pharmaceutical composition of the invention.
  • the device may be, for example, a syringe or an inhaler.
  • compositions of the invention are preferably immunogenic compositions, in that they comprise an immunologically effective amount of an antigen.
  • immunologically effective amount it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention. This amount varies depending upon the health and physical condition of the individual to be treated, age, the taxonomic group of individual to be treated ⁇ e.g. non-human primate, primate, etc.), the capacity of the individual's immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials. Dosage treatment may be a single dose schedule or a multiple dose schedule (e.g. including booster doses). The composition may be administered in conjunction with other immunoregulatory agents.
  • compositions of the invention can be administered directly to the subject.
  • the subjects to be treated can be animals; in particular, human subjects can be treated.
  • Immunogenic compositions of the invention may be used therapeutically (i.e. to treat an existing infection) or prophylactically (i.e. to prevent future infection).
  • An immunogenic composition may include a further adjuvant, which can function to enhance the immune responses (humoral and/or cellular) elicited in a patient who receives the composition.
  • Adjuvants that can be used with the invention include, but are not limited to:
  • a mineral-containing composition including calcium salts and aluminum salts (or mixtures thereof).
  • Calcium salts include calcium phosphate (e.g. the "CAP" particles disclosed in ref. 201).
  • Aluminum salts include hydroxides, phosphates, sulfates, etc., with the salts taking any suitable form (e.g. gel, crystalline, amorphous, etc.). Adsorption to these salts is preferred.
  • the mineral containing compositions may also be formulated as a particle of metal salt [202] .
  • the adjuvants known as aluminum hydroxide and aluminum phosphate may be used. These names are conventional, but are used for convenience only, as neither is a precise description of the actual chemical compound which is present (e.g. see chapter 9 of reference 285).
  • the invention can use any of the "hydroxide” or "phosphate” adjuvants that are in general use as adjuvants.
  • the adjuvants known as "aluminium hydroxide” are typically aluminium oxyhydroxide salts, which are usually at least partially crystalline.
  • the adjuvants known as "aluminium phosphate” are typically aluminium hydroxyphosphates, often also containing a small amount of sulfate (i.e. aluminium hydroxyphosphate sulfate). They may be obtained by precipitation, and the reaction conditions and concentrations during precipitation influence the degree of substitution of phosphate for hydroxyl in the salt.
  • the invention can use a mixture of both an aluminium hydroxide and an aluminium phosphate.
  • aluminium phosphate there may be more aluminium phosphate than hydroxide e.g. a weight ratio of at least 2: 1 e.g. >5: 1, >6: 1, >7: 1, >8: 1, >9: 1, etc.
  • concentration of Al +++ in a composition for administration to a patient is preferably less than lOmg/ml e.g. ⁇ 5 mg/ml, ⁇ 4 mg/ml, ⁇ 3 mg/ml, ⁇ 2 mg/ml, ⁇ 1 mg/ml, etc.
  • a preferred range is between 0.3 and lmg/ml.
  • a maximum of 0.85mg/dose is preferred.
  • Saponins [chapter 22 of ref. 285], which are a heterologous group of sterol glycosides and triterpenoid glycosides that are found in the bark, leaves, stems, roots and even flowers of a wide range of plant species. Saponin from the bark of the Quillaia saponaria Molina tree have been widely studied as adjuvants. Saponin can also be commercially obtained from Smilax ornata (sarsaprilla), Gypsophilla paniculata (brides veil), and Saponaria officianalis (soap root). Saponin adjuvant formulations include purified formulations, such as QS21, as well as lipid formulations, such as ISCOMs.
  • QS21 is marketed as StimulonTM.
  • Saponin compositions have been purified using HPLC and RP-HPLC. Specific purified fractions using these techniques have been identified, including QS7, QS17, QS18, QS21, QH-A, QH- B and QH-C.
  • the saponin is QS21.
  • a method of production of QS21 is disclosed in ref. 203.
  • Saponin formulations may also comprise a sterol, such as cholesterol [204]. Combinations of saponins and cholesterols can be used to form unique particles called immunostimulating complexs (ISCOMs) [chapter 23 of ref. 285].
  • ISCOMs immunostimulating complexs
  • ISCOMs typically also include a phospholipid such as phosphatidyl ethanolamine or phosphatidylcholine. Any known saponin can be used in ISCOMs.
  • the ISCOM includes one or more of QuilA, QHA & QHC.
  • ISCOMs are further described in refs. 204-206.
  • the ISCOMS may be devoid of additional detergent [207].
  • a review of the development of saponin based adjuvants can be found in refs. 208 & 209.
  • Bacterial ADP-ribosylating toxins e.g. the Escherichia coli heat labile enterotoxin "LT”, cholera toxin “CT”, or pertussis toxin “PT”
  • LT heat labile enterotoxin
  • CT cholera toxin
  • PT pertussis toxin
  • the use of detoxified ADP-ribosylating toxins as mucosal adjuvants is described in ref. 21 1 and as parenteral adjuvants in ref. 212.
  • Bioadhesives and mucoadhesives such as esterified hyaluronic acid microspheres [213] or chitosan and its derivatives [214].
  • Microparticles i.e. a particle of -lOOnm to ⁇ 150 ⁇ in diameter, more preferably ⁇ 200nm to ⁇ 30 ⁇ in diameter, or ⁇ 500nm to - ⁇ in diameter
  • materials that are biodegradable and non-toxic e.g. a poly(a-hydroxy acid), a polyhydroxybutyric acid, a polyorthoester, a polyanhydride, a polycaprolactone, etc.
  • poly(lactide-co-glycolide) being preferred, optionally treated to have a negatively-charged surface (e.g. with SDS) or a positively-charged surface (e.g. with a cationic detergent, such as CTAB).
  • a negatively-charged surface e.g. with SDS
  • a positively-charged surface e.g. with a cationic detergent, such as CTAB
  • Liposomes Chosomes 13 & 14 of ref. 285). Examples of liposome formulations suitable for use as adjuvants are described in refs. 215-217.
  • Muramyl peptides such as N-acetylmuramyl-L-threonyl-D-isoglutamine (“thr-MDP"), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N-acetylglucsaminyl-N- acetylmuramyl-L-Al-D-isoglu-L-Ala-dipalmitoxy propylamide ("DTP-DPP", or "TheramideTM), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-( 1 '-2'dipalmitoyl- sn-glycero-3-hydroxyphosphoryloxy)-ethylamine (“MTP-PE").
  • thr-MDP N-acetylmuramyl-L-threonyl-D-isoglutamine
  • a polyoxidonium polymer [218,219] or other N-oxidized polyethylene-piperazine derivative.
  • Methyl inosine 5'-monophosphate (MIMP) [220].
  • a polyhydroxlated pyrrolizidine compound [221] such as one having formula:
  • R is selected from the group comprising hydrogen, straight or branched, unsubstituted or substituted, saturated or unsaturated acyl, alkyl (e.g. cycloalkyl), alkenyl, alkynyl and aryl groups, or a pharmaceutically acceptable salt or derivative thereof.
  • alkyl e.g. cycloalkyl
  • alkenyl alkynyl and aryl groups
  • a pharmaceutically acceptable salt or derivative thereof examples include, but are not limited to: casuarine, casuarine-6-a-D-glucopyranose, 3-epz-casuarine, 7-epz-casuarine, 3,7-diepz-casuarine, etc.
  • a CD Id ligand such as an a-glycosylceramide [222-229] (e.g.
  • a-galactosylceramide phytosphingosine-containing a-glycosylceramides, OCH, KRN7000 [(2S,3S,4R)-l-0-(a-D- galactopyranosyl)-2-(N-hexacosanoylamino)-l ,3,4-octadecanetriol], CRONY- 101 , 3"-0- sulfo-galactosylceramide, etc.
  • An oil-in-water emulsion typically include at least one oil and at least one surfactant, with the oil(s) and surfactant(s) being biodegradable (metabolisable) and biocompatible.
  • the oil droplets in the emulsion are generally less than 5 ⁇ in diameter, and may even have a sub-micron diameter, with these small sizes being achieved with a microfluidiser to provide stable emulsions. Droplets with a size less than 220nm are preferred as they can be subjected to filter sterilization.
  • An immunostimulatory oligonucleotide such as one containing a CpG motif (a dinucleotide sequence containing an unmethylated cytosine residue linked by a phosphate bond to a guanosine residue), or a Cpl motif (a dinucleotide sequence containing cytosine linked to inosine), or a double-stranded RNA, or an oligonucleotide containing a palindromic sequence, or an oligonucleotide containing a poly(dG) sequence.
  • Immunostimulatory oligonucleotides can include nucleotide modifications/analogs such as phosphorothioate modifications and can be double-stranded or (except for RNA) single-stranded.
  • References 231 , 232 and 233 disclose possible analog substitutions e.g. replacement of guanosine with 2'-deoxy-7-deazaguanosine.
  • the adjuvant effect of CpG oligonucleotides is further discussed in refs. 234-239.
  • a CpG sequence may be directed to TLR9, such as the motif GTCGTT or TTCGTT [240].
  • the CpG sequence may be specific for inducing a Thl immune response, such as a CpG-A ODN (oligodeoxynucleotide), or it may be more specific for inducing a B cell response, such a CpG-B ODN.
  • CpG-A and CpG-B ODNs are discussed in refs. 241-243.
  • the CpG is a CpG-A ODN.
  • the CpG oligonucleotide is constructed so that the 5' end is accessible for receptor recognition.
  • two CpG oligonucleotide sequences may be attached at their 3' ends to form "immunomers". See, for example, references 240 & 244-246.
  • CpG7909 also known as ProMuneTM (Coley Pharmaceutical Group, Inc.). Another is CpG1826.
  • TpG sequences can be used [247], and these oligonucleotides may be free from unmethylated CpG motifs.
  • the immunostimulatory oligonucleotide may be pyrimidine-rich. For example, it may comprise more than one consecutive thymidine nucleotide (e.g. TTTT, as disclosed in ref. 247), and/or it may have a nucleotide composition with >25% thymidine (e.g.
  • oligonucleotides may be free from unmethylated CpG motifs.
  • Immunostimulatory oligonucleotides will typically comprise at least 20 nucleotides. They may comprise fewer than 100 nucleotides.
  • an adjuvant used with the invention may comprise a mixture of (i) an oligonucleotide (e.g. between 15-40 nucleotides) including at least one (and preferably multiple) Cpl motifs, and (ii) a polycationic polymer, such as an oligopeptide (e.g. between 5-20 amino acids) including at least one (and preferably multiple) Lys-Arg-Lys tripeptide sequence(s).
  • the oligonucleotide may be a deoxynucleotide comprising 26-mer sequence 5'-(IC) i3-3' (SEQ ID NO:98).
  • the polycationic polymer may be a peptide comprising 1 1-mer amino acid sequence KLKLLLLLKLK (SEQ ID NO: 99).
  • 3dMPL 3-O-deacylated monophosphoryl lipid A
  • '3dMPL also known as 'MPLTM'
  • 3dMPL can form micellar aggregates or particles with different sizes e.g. with a diameter ⁇ 150nm or >500nm. Either or both of these can be used with the invention, and the better particles can be selected by routine assay. Smaller particles (e.g. small enough to give a clear aqueous suspension of 3dMPL) are preferred for use according to the invention because of their superior activity [253].
  • Preferred particles have a mean diameter less than 220nm, more preferably less than 200nm or less than 150nm or less than 120nm, and can even have a mean diameter less than lOOnm. In most cases, however, the mean diameter will not be lower than 50nm.
  • An imidazoquinoline compound such as Imiquimod (“R-837”) [254,255], Resiquimod (“R-848”) [256], and their analogs; and salts thereof (e.g. the hydrochloride salts). Further details about immunostimulatory imidazoquinolines can be found in references 257 to 261.
  • a thiosemicarbazone compound such as those disclosed in reference 262. Methods of formulating, manufacturing, and screening for active compounds are also described in reference 262.
  • the thiosemicarbazones are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF-a.
  • a tryptanthrin compound such as those disclosed in reference 263.
  • Methods of formulating, manufacturing, and screening for active compounds are also described in reference 263.
  • the thiosemicarbazones are particularly effective in the stimulation of human peripheral blood mononuclear cells for the production of cytokines, such as TNF-a.
  • a nucleoside analog such as: (a) Isatorabine (ANA-245; 7-thia-8-oxoguanosine):
  • a phosphazene such as poly[di(carboxylatophenoxy)phosphazene] ("PCPP") as described, for example, in references 275 and 276.
  • PCPP poly[di(carboxylatophenoxy)phosphazene]
  • Antigens and adjuvants in a composition will typically be in admixture.
  • Compositions may include two or more of said adjuvants.
  • they may advantageously include both an oil-in-water emulsion and 3dMPL, etc.
  • oil-in-water emulsion adjuvants useful with the invention include, but are not limited to:
  • a submicron emulsion of squalene, Tween 80, and Span 85 A submicron emulsion of squalene, Tween 80, and Span 85.
  • the composition of the emulsion by volume can be about 5% squalene, about 0.5% polysorbate 80 and about 0.5% Span 85. In weight terms, these ratios become 4.3% squalene, 0.5% polysorbate 80 and 0.48% Span 85.
  • This adjuvant is known as 'MF59®' [282-284], as described in more detail in Chapter 10 of ref. 285 and chapter 12 of ref. 286.
  • the MF59® emulsion advantageously includes citrate ions e.g. lOmM sodium citrate buffer.
  • An emulsion of squalene, a tocopherol, and Tween 80 may include phosphate buffered saline. It may also include Span 85 ⁇ e.g. at 1%) and/or lecithin. These emulsions may have from 2 to 10%) squalene, from 2 to 10%) tocopherol and from 0.3 to 3% Tween 80, and the weight ratio of squalene:tocopherol is preferably ⁇ 1 as this provides a more stable emulsion. Squalene and Tween 80 may be present volume ratio of about 5:2.
  • One such emulsion can be made by dissolving Tween 80 in PBS to give a 2% solution, then mixing 90ml of this solution with a mixture of (5g of DL-a-tocopherol and 5ml squalene), then micro fluidising the mixture.
  • the resulting emulsion may have submicron oil droplets e.g. with an average diameter of between 100 and 250nm, preferably about 180nm.
  • An emulsion of squalene, a tocopherol, and a Triton detergent e.g. Triton X-100.
  • the emulsion may also include a 3d-MPL (see below).
  • the emulsion may contain a phosphate buffer.
  • An emulsion comprising a polysorbate (e.g. polysorbate 80), a Triton detergent (e.g. Triton X-100) and a tocopherol (e.g. an a-tocopherol succinate).
  • the emulsion may include these three components at a mass ratio of about 75: 1 1 : 10 (e.g. 750 ⁇ g/ml polysorbate 80, 1 10 ⁇ g/ml Triton X-100 and 100 ⁇ g/ml a-tocopherol succinate), and these concentrations should include any contribution of these components from antigens.
  • the emulsion may also include squalene.
  • the emulsion may also include a 3d-MPL (see below).
  • the aqueous phase may contain a phosphate buffer.
  • An emulsion of squalane, polysorbate 80 and poloxamer 401 (“PluronicTM L121").
  • the emulsion can be formulated in phosphate buffered saline, pH 7.4.
  • This emulsion is a useful delivery vehicle for muramyl dipeptides, and has been used with threonyl-MDP in the "SAF-1" adjuvant [287] (0.05-1% Thr-MDP, 5% squalane, 2.5% Pluronic L121 and 0.2% polysorbate 80). It can also be used without the Thr-MDP, as in the "AF” adjuvant [288] (5% squalane, 1.25% Pluronic L121 and 0.2% polysorbate 80). Microfluidisation is preferred.
  • An emulsion having from 0.5-50%) of an oil, 0.1-10%) of a phospholipid, and 0.05-5%o of a non-ionic surfactant.
  • preferred phospholipid components are phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidylglycerol, phosphatidic acid, sphingomyelin and cardiolipin. Submicron droplet sizes are advantageous.
  • Additives may be included, such as QuilA saponin, cholesterol, a saponin-lipophile conjugate (such as GPI-0100, described in reference 290, produced by addition of aliphatic amine to desacylsaponin via the carboxyl group of glucuronic acid), dimethyidioctadecylammonium bromide and/or NN-dioctadecyl- NN-bis (2-hydroxyethyl)propanediamine .
  • a non-metabolisable oil such as light mineral oil
  • surfactant such as lecithin, Tween 80 or Span 80.
  • Additives may be included, such as QuilA saponin, cholesterol, a saponin-lipophile conjugate (such as GPI-01
  • compositions of the invention include MF59®, Alum and/or toll-like receptor 7 (TLR7) agonists, for example, as disclosed in EP2459216 herein incorporated by reference.
  • TLR7 toll-like receptor 7
  • the invention also provides a conjugate of the invention, for use in medicine e.g. for use in raising an antibody response in a suitable mammal.
  • the invention also provides a method for raising an immune response in a suitable mammal, comprising administering a conjugate or pharmaceutical composition of the invention to the mammal.
  • the invention also provides the use of a conjugate of the invention in the manufacture of a medicament for preventing or treating a microbial infection in a suitable mammal.
  • the immune response raised by these methods and uses will generally include an antibody response, preferably a protective antibody response. Methods for assessing antibody responses after antigen immunisation are well known in the art.
  • the antibody response is preferably an IgA or IgG response.
  • the immune response may be prophylactic and/or therapeutic.
  • the mammal is preferably a human, particularly a human female, more particularly a pregnant human female.
  • Efficacy of therapeutic treatment can be tested by monitoring microbial infection after administration of the composition of the invention.
  • Efficacy of prophylactic treatment can be tested by monitoring immune responses against antigen (e.g. anti-antigen antibodies) after administration of the composition.
  • antigen e.g. anti-antigen antibodies
  • compositions of the invention will generally be administered directly to a patient.
  • Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or by rectal, oral, vaginal, topical, transdermal, intradermal, ocular, nasal, aural, or pulmonary administration. Injection or intranasal administration is preferred.
  • the invention may be used to elicit systemic and/or mucosal immunity.
  • Vaccines prepared according to the invention may be used to treat both children and adults.
  • a subject may be less than 1 year old, 1-5 years old, 5-15 years old, 15-55 years old, or at least 55 years old.
  • Particular subjects for receiving the vaccines are the elderly (e.g. >50 years old, >55 years old, >60 years old, and >65 years), or the young (e.g. ⁇ 5 years old).
  • the vaccines are not suitable solely for these groups, however, and may be used more generally in a population.
  • Treatment can be by a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primary immunisation schedule and/or in a booster immunisation schedule. In a multiple dose schedule the various doses may be given by the same or different routes e.g. a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc. Administration of more than one dose (typically two doses) is particularly useful in immunologically naive patients. Multiple doses will typically be administered at least 1 week apart (e.g. about 2 weeks, about 3 weeks, about 4 weeks, about 6 weeks, about 8 weeks, about 10 weeks, about 12 weeks, about 16 weeks, etc.).
  • the uses and methods of the invention are particularly useful for treating/protecting against infections caused by the organism from which the antigen is derived. Exemplary uses/methods are discussed below.
  • the uses and methods may be for the prevention and/or treatment of a disease caused by Streptococcus agalactiae.
  • the examples demonstrate that the carrier molecules of the invention are particularly effective carriers for S. agalactiae saccharides.
  • the BP-2a and spbl polypeptides of the carrier may also be able to help induce a carrier-specific immune response that is protective.
  • Immunogenic compositions comprising carrier proteins of the invention may be for use in: (i) a method for treating or preventing an infectious disease caused by Streptococcus agalactiae; or (ii) a method for inducing an immune response against Streptococcus agalactiae in a subject; or (iii) a method of vaccinating a subject to protect against or reduce the symptoms of infection by Streptococcus agalactiae, wherein the method comprises administering to a subject in need thereof a therapeutically effective amount of said composition.
  • immunogenic compositions comprising carrier proteins of the invention may be for use in a method of providing protective immunity against Streptococcus agalactiae or protecting from a disease caused by Streptococcus agalactiae in an infant, wherein the infant was born to a female to whom a therapeutically effective amount of the composition was administered during the time when said female was pregnant with said infant.
  • the disease caused by Streptococcus agalactiae may be Early Onset Disease (EOD) and/or Late Onset Disease (LOD).
  • the uses and methods may be for the prevention and/or treatment of a disease caused by Neisseria meningitidis, e.g. meningitis, septicaemia, etc.
  • anti-glucan immunity may have efficacy against a broad range of pathogens and diseases.
  • anti-glucan serum raised after immunisation with Saccharomyces cerevisiae is cross-reactive with Candida albicans.
  • Broad spectrum immunity is particularly useful because, for these human infectious fungal agents, chemotherapy is scanty, antifungal drug resistance is emerging and the need for preventative and therapeutic vaccines is increasingly recognized.
  • Candida species such as C. albicans
  • Cryptococcus species such as C. neoformans
  • Enterococcus species such as E. faecalis
  • Streptococcus species such as S. pneumoniae, S. mutans, S. agalactiae and S. pyogenes
  • Leishmania species such as L. major
  • Acanthamoeba species such as A. castellani
  • Aspergillus species such as A. fumigatus and A.flavus
  • Pneumocystis species such as P. carinii
  • Mycobacterium species such as M.
  • tuberculosis Pseudomonas species, such as P. aeruginosa; Staphylococcus species, such as S. aureus; Salmonella species, such as S. typhimurium; Coccidioides species such as C. immitis; Trichophyton species such as T. verrucosum; Blastomyces species such as B. dermatidis; Histoplasma species such as H. capsulatum; Paracoccidioides species such as P. brasiliensis; Pythium species such as P. insidiosum; and Escherichia species, such as E. coli.
  • the uses and methods are particularly useful for preventing/treating diseases including, but not limited to: candidiasis (including hepatosplenic candidiasis, invasive candidiasis, chronic mucocutaneous candidiasis and disseminated candidiasis); candidemia; aspergillosis, cryptococcosis, dermatomycoses, sporothrychosis and other subcutaneous mycoses, blastomycosis, histoplasmosis, coccidiomycosis, paracoccidiomycosis, pneumocystosis, thrush, tuberculosis, mycobacteriosis, respiratory infections, scarlet fever, pneumonia, impetigo, rheumatic fever, sepsis, septicaemia, cutaneous and visceral leishmaniasis, corneal acanthamoebiasis, cystic fibrosis, typhoid fever, gastroenteritis and hemolytic-uremic syndrome.
  • Conjugates of the invention may be combined with non-glucan antigens into a single composition for simultaneous immunisation against multiple pathogens.
  • conjugates may be administered to patients at substantially the same time as ⁇ e.g. during the same medical consultation or visit to a healthcare professional or vaccination centre) other vaccines.
  • Antigens for use in these combination vaccines or for concomitant administration include, for instance, immunogens from Streptococcus agalactiae, Staphylococcus aureus and/or Pseudomonas aeuruginosa, hepatitis A virus, hepatitis B virus, Neisseria meningitidis (such as saccharides or conjugated saccharides, for serogroups A, C, W135 and/or Y), Streptococcus pneumoniae (such as saccharides or conjugated saccharides), etc.
  • Conjugates of the invention may be used in conjunction with anti-fungals, particularly where a patient is already infected.
  • the anti-fungal offers an immediate therapeutic effect whereas the conjugate offers a longer-lasting effect.
  • Suitable anti-fungals include, but are not limited to, azoles (e.g. fluconazole, itraconazole), polyenes (e.g. amphotericin B), flucytosine, and squalene epoxidase inhibitors (e.g. terbinafine) [see also ref. 292].
  • the anti-fungal and the conjugate may be administered separately or in combination. When administered separately, they will typically be administered within 7 days of each other. After the first administration of an conjugate, the antifungal may be administered more than once.
  • the uses and methods may be for the prevention and/or treatment of a disease caused by Group A streptococcus.
  • the uses and methods may be for the prevention and/or treatment of a disease caused by pneumococcus, e.g. meningitis, sepsis, pneumonia etc.
  • GI numbering is used above.
  • a GI number, or “Genlnfo Identifier” is a series of digits assigned consecutively to each sequence record processed by NCBI when sequences are added to its databases. The GI number bears no resemblance to the accession number of the sequence record.
  • a sequence is updated ⁇ e.g. for correction, or to add more annotation or information) then it receives a new GI number.
  • sequence associated with a given GI number is never changed.
  • an antigen "domain” is omitted, this may involve omission of a signal peptide, of a cytoplasmic domain, of a transmembrane domain, of an extracellular domain, etc.
  • compositions comprising
  • consisting encompasses “including” as well as “consisting” e.g. a composition "comprising” X may consist exclusively of X or may include something additional e.g. X + Y.
  • consisting essentially of means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
  • Consisting of is generally taken to mean that the invention as claimed is limited to those elements specifically recited in the claim (and may include their equivalents, insofar as the doctrine of equivalents is applicable).
  • in relation to a numerical value x means, for example, + two standard deviations of the value. In certain embodiments, "about” is understood as acceptable variation and tolerances within the specific art. For example, when referring to a measurable value such as an amount, a temporal duration, and the like, may encompass variations of ⁇ 20% or ⁇ 10%, more preferably ⁇ 5%, even more preferably ⁇ 1%, and still more preferably ⁇ 0.1%) from the specified value, as such variations are appropriate to perform the disclosed methods.
  • substantially does not exclude “completely” e.g. a composition which is “substantially free” from Y may be completely free from Y.
  • “substantially free” from Y can be understood as a composition containing not more than 5%> Y, not more than 4%> Y, not more than 3%o Y, not more than 2%> Y, not more than 1%> Y, or not more than 0.1% Y.
  • the word “substantially” may be omitted from the definition of the invention.
  • canonical in relation to amino acids means that the amino acid is one of the twenty amino acids encoded by the universal genetic code, i.e. Alanine, Asparagine, Aspartic acid, Arginine, Cysteine, Glutamine, Glycine, Glutamic acid, Histidine, Isoleucine, Lysine, Leucine, Phenylalanine, Methionine, Serine, Proline, Tryptophan, Threonine, Tyrosine and Valine.
  • fragment refers to a sequence that is a subset of another sequence.
  • fragment and “subsequence” are used interchangeably. These terms are used to refer to a part or portion of an intact or complete wild-type polypeptide but which comprise fewer amino acid residues than an intact or complete wild- type polypeptide. Thus, the term refers to truncated or shorter amino acid sequences corresponding to one or more regions of a wild-type or reference polypeptide.
  • fragments are truncated or shorter fragments of a reference sequence
  • fragments may be modified to comprise additional sequences not found in the reference polypeptide, for example, to form fusion polypeptides, include 'tag' sequences such as His tags or Glutathione S- transferase (GST) tags, linker sequences and the like.
  • 'tag' sequences such as His tags or Glutathione S- transferase (GST) tags, linker sequences and the like.
  • the amino group of the N terminal amino acid of the fragment is not linked by a peptide bond to the carboxyl group of an amino acid to which it is linked in the reference polypeptide and/or the carboxyl group of the C terminal amino acid of the fragment is not linked by a peptide bond to the amino group of an amino acid to which it is linked in the reference polypeptide.
  • the percent identity of a first polypeptide and a second polypeptide is generally determined by counting the number of matched positions between the first and second polypeptides and dividing that number by the total length of the shortest polypeptide followed by multiplying the resulting value by 100. For fragments of polypeptides this value is usually determined to be around 100% and therefore has little meaning.
  • Proportion of reference polypeptide (expressed as a percentage) may be used. Proportion of reference polypeptide is calculated by counting the number of matched positions between the fragment and reference polypeptides and dividing that number by the total length of the reference polypeptide followed by multiplying the resulting value by 100. Particularly, fragments will comprise less than 90, 80, 70, 65, 60, 55, 50, 45, 40, 35, 30, 25 or less than 20% of the sequence of the reference polypeptide.
  • a process comprising a step of mixing two or more components does not require any specific order of mixing.
  • components can be mixed in any order. Where there are three components then two components can be combined with each other, and then the combination may be combined with the third component, etc.
  • animal (and particularly bovine) materials are used in the culture of cells, they should be obtained from sources that are free from transmissible spongiform encaphalopathies (TSEs), and in particular free from bovine spongiform encephalopathy (BSE). Overall, it is preferred to culture cells in the total absence of animal-derived materials.
  • TSEs transmissible spongiform encaphalopathies
  • BSE bovine spongiform encephalopathy
  • a compound is administered to the body as part of a composition then that compound may alternatively be replaced by a suitable prodrug.
  • references to a percentage sequence identity between two amino acid sequences means that, when aligned, that percentage of amino acids are the same in comparing the two sequences. Generally, percentage sequence identity is calculated by dividing the number of identical amino acids by the total number of amino acids in the longer sequence.
  • This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of ref. 300.
  • a preferred alignment is determined by the Smith- Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62.
  • the Smith- Waterman homology search algorithm is disclosed in ref. 301.
  • the fusion protein "GBS59(6XD3)-GSG4-1523" gene was generated by PIPE cloning by amplifying the gene of 59(6XD3) (again from a synthetic gene purchased from geneart) using the primers 59pipeF (SEQ ID NO:278), 59GSG41523.R (SEQ ID NO:281) and, the gbsl523 gene from genomic GBS COHI strain DNA using the primers 59GSG41523.F (SEQ ID NO:280) and 1523pipe.R (SEQ ID NO:279).
  • the two I-PCR containing an overlapping region of 18 bp (SEQ ID NO:282) corresponding to the GSG4 linker (SEQ ID NO:272), were co-used in a PIPE reaction using the pET15 TEV Vector-PCR (coding for N-terminal 6XHIS-TEV tag ("6XHIS" disclosed as SEQ ID NOs: 79 and 275)) and co-transformed in the HK100 E.coli Strain. The correct sequence was verified by DNA sequencing of the obtained clones.
  • the 59(6XD3)-GSG4-1523 gene was further amplified from the pET15-TEV-59(6XD3)-GSG4-1523 clone ("GSG4" disclosed as SEQ ID NO: 272) by using the primers 596xD3 Nhel F (SEQ ID NO:283) and 1523 Xhol Stop R (SEQ ID NO:284).
  • the PCR product was cleaved with NheLXhoI restriction enzymes and ligated in to the pET24b+ vector (novagen) previously digested with the same restriction enzymes.
  • the ligation reaction was transformed in DH5a E. coli cells and the correct sequence of the clones was verified by DNA sequencing.
  • the pET24-59(6XD3)-GSG4-1523 clone (kanamycin resistance) was transformed in BL21(DE3) tl resistant strain (NEB). Protein expression was achieved by inoculating one single colony in LB-PTK + kanamycin medium and inducing protein expression by the addition of IPTG.
  • the cultures were maintained at 25°C for 5h after induction with ImM IPTG for the pET clones. All recombinant proteins were purified by affinity chromatography and gel filtration. Briefly, cells were harvested by centrifugation and lysed in "lysis buffer", containing lOmM imidazole, lmg ⁇ ml lysozyme, 0.5 mg ⁇ ml DNAse and COMPLETE inhibitors cocktail (Roche) in PBS. The lysate was clarified by centrifugation and applied onto His- Trap HP column (Armesham Biosciences) pre-equilibrated in PBS containing lOmM imidazole.
  • the GBS59(6XD3)-GBS1523-HIS fusion was tested for its ability to provide protection against a range of GBS challenge strains. Separate groups of mice were immunised with the GBS59(6XD3)-GBS1523-HIS fusion and were then challenged with different GBS challenge strains that express different GBS59 alleles, or express GBS1523. Immunisation with PBS was used as a negative control. As shown in Table 5, the presence of GBS59 and GBS 1523 antigens in the fusion provided protection against strains that express GBS59, and against strains that express GBS 1523.
  • the fusion comprising GBS 1523 provided protection comparable to that obtained with the GBS59(6XD3) fusion disclosed in WO2011/121576 against strains expressing different GBS59 alleles demonstrating that addition of GBS 1523 does not negatively impact immunogenicity.
  • the efficacy of the GBS59(6XD3)-GBS1523 conjugate as a carrier for GBS type II polysaccharide was compared to other carrier proteins. Groups of mice were administered three doses of the relevant saccharide conjugate in Alum at a dose of ⁇ g protein per dose. As shown in Table 6, the GBS59(6XD3)-GBS1523 conjugate is a particularly effective carrier for GBS type II polysaccharide and 100% of mice immunised with GBS59(6XD3)-GBS 1523-11 survived challenge with GBS type II strain 5401.
  • GBS59(6XD3)-GBS 1523-11 was greater than the protection provided by CRM-II (85%) survival) and greater than the protection provided by GBS80-II (78%o survival).
  • GBS59(6XD3)-GBS 1523-11 provided good protection against challenge by type III strain COH1 (100%> survival) and type la strain 515 (40%> survival).
  • the titre of antibodies specific for type II saccharide elicited by the compositions are shown in Figure 1.
  • GBS59(6XD3)-GBS 1523-11 elicited high titres of specific antibodies and the titres are comparable to or higher than the titres elicited by CRM-II. Again, these data are significant, because they indicate that the increased protection exhibited by GBS59(6XD3)-GBS 1523-11 relative to, for example, CRM-II is not merely a result of the presence of carrier proteins that comprise GBS antigens.
  • the GBS59(6XD3)-GBS 1523 conjugate is effective as a carrier molecule and enhances the immune response that is specific for the type II saccharide antigen.
  • the titre of antibodies specific for GBS59 and GBS1523 elicited by the GBS59(6XD3)-GBS1523-II compositions are shown in Figure 2.
  • the efficacy of the GBS59(6XD3)-GBS1523 conjugate as a carrier for GBS type V polysaccharide was compared to other carrier proteins. Groups of mice were administered three doses of the relevant saccharide conjugate in Alum at a dose of ⁇ g protein per dose. As shown in Table 7, the GBS59(6XD3)-GBS1523 conjugate is a particularly effective carrier for GBS type V polysaccharide and 86% of mice immunised with GBS59(6XD3)-GBS 1523-V survived challenge with GBS type V strain CJB1 1 1. In addition, GBS59(6XD3)-GBS 1523-V provided good protection against challenge by type III strain COH1 (60% survival).
  • the efficacy of the GBS59(6XD3)-GBS 1523 conjugate as a carrier for GBS type II and type V polysaccharides was also compared to TT, CRM197 and GBS80.
  • Groups of 8 mice were administered three doses of the relevant saccharide conjugate in Alum at a dose of ⁇ g saccharide and 400 ⁇ g Alum per dose
  • immunisation with GBS59(6XD3)-GBS 1523-11 provided greater protection against challenge with type II strain 5401 than CRM-II and GBS80-II, and also greater protection than TT-II.
  • Figure 3 provides the geometric mean titres of anti-II saccharide antibodies elicited by the different conjugates in this study.
  • the titres elicited by GBS59(6XD3)-GBS 1523-11 were greater than CRM-II and GBS80-II. These data are significant, because they indicate that the increased protection exhibited by GBS59(6XD3)-GBS 1523-11 relative to, for example, CRM-II is not merely a result of the presence of carrier proteins that comprise GBS antigens.
  • the GBS59(6XD3)- GBS1523 conjugate is effective as a carrier molecule and enhances the immune response that is specific for the type II saccharide antigen.
  • Figure 4 provides the geometric mean titres of anti-V saccharide antibodies elicited by the different conjugates in this study.
  • the titres elicited by GBS59(6XD3)-GBS 1523-V were greater than CRM-V and GBS80-V. These data are significant, because they indicate that the increased protection exhibited by GBS59(6XD3)-GBS1523-V relative to, for example, CRM-V is not merely a result of the presence of carrier proteins that comprise GBS antigens.
  • the GBS59(6XD3)- GBS1523 conjugate is effective as a carrier molecule and enhances the immune response that is specific for the type V saccharide antigen.
  • the titre of antibodies specific for type V saccharide elicited by the compositions tested are shown in Figure 5.
  • GBS59(6XD3)-GBS1523-V elicited high titres of specific antibodies and the titres are comparable or higher than the titres elicited by CRM-V.
  • these data are significant, because they indicate that the increased protection exhibited by GBS59(6XD3)-GBS1523-V relative to, for example, CRM-V is not merely a result of the presence of carrier proteins that comprise GBS antigens.
  • the GBS59(6XD3)-GBS1523 conjugate is effective as a carrier molecule and enhances the immune response that is specific for the type V saccharide antigen.
  • GBS proteins The ability of other GBS proteins to function as carrier proteins was tested using Laminarin as a capsular polysaccharide model.
  • GBS52 Only one protein (GBS52) was able to function as a carrier protein eliciting anti-laminarin IgG titers comparable to those of the laminarin-CRM197 conjugate used as a benchmark ( Figure 7).
  • the GBS59(6XD3)-GBS1523 conjugate was tested for its efficacy when used in combination with other carrier-saccharide conjugates.
  • GBS59(6XD3)-GBS 1523-11 and GBS59(6XD3)- GBS1523-V conjugates were combined with a trivalent vaccine comprising la, lb and III GBS saccharides conjugated to CRM, and with GBS80 conjugates.
  • the protection provided by these tetravalent and pentavalent compositions was compared to compositions comprising the trivalent vaccine in combination with serotype-II and -V saccharides conjugated to CRMi 97 and GBS80.
  • Tables 9 and 10 demonstrate the efficacy of the GBS59(6XD3)-GBS1523 conjugate as a carrier for both type -II and -V saccharides, when administered alone or in combination with other saccharide conjugates.
  • GBS59(6XD3)-GBS 1523-11 provides better protection against serotype-II strain 5401 than CRM-II and GBS80-II when the conjugates are administered individually (90% survival compared to 67% and 69%).
  • the improved protection against serotype-II strain 5401 is maintained when GBS59(6XD3)-GBS 1523-11 conjugates are combined with the trivalent CRM-Ia/Ib/III composition.
  • a tetravalent composition comprising GBS59(6XD3)-GBS 1523-11 and CRM-Ia/Ib/III provides higher protection than a composition comprising CRM-II and CRM-Ia/Ib/III (81 ) survival compared to 73% survival). This protection is increased further when GBS80-V is added to generate a pentavalent composition (96% survival).
  • GBS59(6XD3)-GBS1523-V provides strong protection against serotype-V strain CJB 1 1 1 that is comparable to the protection provided by CRM-V.
  • GBS59(6XD3)-GBS1523-V is also effective when combined with the trivalent CRM-Ia/Ib/III composition to generate tetravalent and pentavalent compositions.
  • the GBS59(6XD3)-GBS 1523-11 and GBS59(6XD3)-GBS1523-V conjugates were also tested for their ability to provide protection against heterologous strains when combined with the trivalent CRM-Ia/Ib/III composition and GBS80 conjugates. As shown in Tables 1 1 and 12 below, although protection provided by the conjugates was modest, it was greater than the protection provided by PBS and the GBS59(6XD3)-GBS1523 and GBS80 proteins themselves.
  • Example 8 GBS59(6XD3)-GBS1523 conjugate as a carrier in multivalent vaccines - antibody titres
  • Figure 8 shows the titres of antibodies specific for type II saccharide elicited by a range of different multivalent vaccine compositions ("Triv.” refers to the CRM-Ia/Ib/III trivalent vaccine).
  • the titre elicited by compositions comprising the GBS59(6XD3)-GBS1523 carrier was greater than that elicited by corresponding compositions comprising CRM- conjugates.
  • polysaccharide interference was not observed for the pentavalent GBS59(6XD3)-GBS1523 carrier compositions.
  • a more dispersed immune response is visible, potentially as a result of carrier immune epitope suppression from the high levels of CRM 197 used in the specific mouse model.
  • Figure 9 shows the titres of antibodies specific for type V saccharide elicited by a range of different multivalent vaccine compositions ("Triv.” refers to the CRM-Ia/Ib/III trivalent vaccine).
  • the titre elicited by compositions comprising the GBS59(6XD3)-GBS1523 carrier was greater than that elicited by corresponding compositions comprising CRM- conjugates.
  • polysaccharide interference was not observed for the pentavalent GBS59(6XD3)-GBS1523 carrier compositions.
  • Figures 10 and 11 provide further statistical analysis of some of the data shown in Figures 8 and 9. It can be seen that the GBS59(6XD3)-GBS1523 carriers were statistically significantly more effective than the CRMi 97 compositions.
  • the combination of three proteins can potentially protect against a broader range of GBS isolates, representing a more efficient solution for the prevention of GBS infections.
  • the fusion protein approach offers substantial advantages: it is a more defined product and it reduces the number of recombinant expression and purification steps, making the difference in terms of costs and feasibility for the vaccine.
  • Fusion of pilus 2a chimeric protein with the pilus 2b backbone protein resulted in a highly expressed and stable antigen in which each domain folds individually correctly.
  • the recombinant chimera, alone or conjugated with GBS polysaccharides, conferred strong protection in mice challenged with any of the GBS strains carrying pilus 2b or pilus 2a variants.
  • Epitopes inducing protective immune responses are restricted to specific domains within an immunogenic protein. Once the domains are identified, they can be expressed in a recombinant form, for use as vaccine antigens devoid of regions that are irrelevant from a vaccine standpoint.
  • One of the advantages of working with protein domains is that they are typically structurally ordered, and can be easily handled in vitro, further simplifying the construction of synthetic fusion proteins hosting two or more immunogenic domains. Therefore, to determine if the spbl (GBS 1523) region of the fusion protein could be further optimised, structural vaccinology was applied to define the minimal domain carrying the protective epitopes.
  • BP-2b 3 o-468 full length spbl
  • BP-2bi 85-468 BP- 2b D 2+D3, SEQ ID NO: 285
  • Dl 30 _i84, D2 185 -356, D3 3 47-468, SEQ ID NOs:287, 289, 291 were PCR amplified from GBS strain COH1 (SAN- 1518), cloned in the pET21b+ vector and expressed as C-terminal His-tagged proteins. Proteins were expressed in E.
  • the soluble proteins were extracted by using the cell lytic sigma reagent solubilized in Tris pH 7.5, 300mM NaCl, lOmM imidazole, and purified by a FF-Crude His-Trap HP nickel chelating column (Amersham Bioscience).
  • the recombinant proteins, eluted with 300mM imidazole, were concentrated by ultrafiltration to 10 mg ⁇ ml and loaded onto HiLoad 26/60 Superdex 75 (Amersham Biosciences) pre-equilibrated in 50mM Tris-HCl (pH 7.5), 75 mM NaCl.
  • the protein abundance in pure fractions was quantified with the BCA assay (Pierce).
  • S. agalactiae strains were grown at 37 °C in 5% C0 2 in Todd Hewitt Broth (Difco Laboratories) or in trypticase soy agar supplemented with 5% sheep blood.
  • mice A mouse maternal immunization/pup challenge model of GBS infection was used to verify the protective efficacy of the antigens, as previously described (Maione et al, 2005).
  • groups of 5 CD-I female mice (6-8 weeks old) were immunized on days 1 , 21 and 35 with either PBS or 20 ⁇ g of protein per dose in Alum formulations.
  • Mice were bred 3 days after the last immunization.
  • pups were injected intraperitoneally with a dose of GBS bacteria (COH1 strain) calculated to cause 90% lethality.
  • Mice were monitored on a daily basis and euthanized when they exhibited defined humane endpoints that had been pre-established for the study in agreement with Novartis Animal Welfare Policies.
  • Statistical analysis was performed using Fisher's exact test.
  • BP-2b D 2+D3 retains the immunological proprieties of full length protein
  • the purified recombinant proteins were independently used to immunize CD1 mice and the protection activity of each was tested in an active maternal mouse immunization/neonatal pup challenge model.
  • the GBS strain COH1 expressing high level of BP-2b protein on its surface.
  • the BP D2+ D3 construct conferred in vivo protection to a level comparable to that of the full length protein, suggesting that protective epitopes in BP-2b are specifically concentrated in this portion of the protein, while the Dl domain appears dispensable for the protection in mice.
  • Figure 14 presents western blotting with serum from human patient which strongly recognizes full length spb l (GBS 1523) and spbl D2 +D3 (GBS1523 D2 +D3) but not spbl D i (GBS 1523 D i).
  • sequence of an exemplary GBS59(6xD3)-GBS 1523 D 2+D3 is provided as SEQ ID:293.
  • sequence of a further exemplary carrier of the invention comprising the seven D3 subdomains of GBS59 and the D2+D3 domains of GBS 1523, GBS59(7XD3 SU B)-GBS 1523 D2+ D3, is provided as SEQ ID:294.
  • Variant sequences of GBS 1523 domains Dl , D2 and D3 are provided in SEQ ID NOs:288, 290 and 292.
  • SEQ ID NO:57 (515 D2+D3+D4H)
  • SEQ ID NO:58 (cjbl 11 D3+D4)
  • SEQ ID NO:99 Polycationic peptide adjuvant
  • SEQ ID NO: 100 (encoding GBS59 2603)
  • SEQ ID NO: 104 (encoding GBS59 CJBl 10)
  • SEQ ID NO: 158 encoding cjbl 11 D3+D4H)
  • SEQ ID NO: 159 encoding cjbl 11 D2+D3+D4)
  • SEQ ID NO: 160 encoding cjbl 11 D2+D3+D4H)
  • SEQ ID NO: 161 encoding h36b D3+D4)
  • SEQ ID NO: 180 (GBS80 2603 without transmembrane/cytoplasmic region and cell wall anchor)
  • SEQ ID NO: 181 GS80 2603 without extracellular domain
  • SEQ ID NO: 182 N-terminal immunogenic fragment of GBS80 2603
  • SEQ ID NO: 186 N-terminal fragment of GBS67 2603
  • SEQ ID NO: 187 N-terminal fragment of GBS67 2603
  • SEQ ID NO: 189 N-terminal fragment of GBS67 h36b
  • SEQ ID NO: 192 N-terminal fragment of GBS67 CJB 1 11
  • SEQ ID NO: 196 N-terminal fragment of GBS67 515)
  • SEQ ID NO:201 N-terminal fragment of GBS67 DK21
  • SEQ ID NO:202 N-terminal fragment of GBS67 D 21
  • SEQ ID NO:204 N-terminal fragment of GBS67 CJB 1 10)
  • SEQ ID NO:205 N-terminal fragment of GBS67 CJB 1 10)
  • SEQ ID NO:217 (GBS3 2603 coiled coil and proline-rich segments)
  • SEQ ID NO:218 (GBS3 2603 signal sequence and coiled coil)
  • SEQ ID NO:220 (GBS3 2603 signal sequence, coiled coil and proline rich segment)
  • SEQ ID NO:270 epitopope of D3 bound by 4H1 1/B7 and 17C4/A3
  • SEQ ID NO:278 (PCR Primer 59pipeF)
  • SEQ ID NO:280 (PCR Primer 59GSG41523.F)
  • SEQ ID NO:281 (PCR Primer 59GSG41523.R)
  • SEQ ID NO:284 (PCR Primer 1523 Xhol Stop R)
  • SEQ ID NO:286 (GBS1523 D2+D3 with aditional N-terminal sequence)
  • SEQ ID NO:293 (GBS59(6xD3)-GBS 1523 D2+D3 Fusion Protein)
  • SEQ ID NO:294 (GBS59(7XD3 SUB )-GBS 1523 D2+D 3 Fusion Protein)
  • SEQ ID NO:295 (GBS59(7XD3 SUB )-GBS 1523 D2+D 3 Fusion Protein)
  • Vaccine Adjuvants Preparation Methods and Research Protocols (Volume 42 of Methods in Molecular Medicine series). ISBN: 1-59259-083-7. Ed. O'Hagan
PCT/EP2015/068012 2014-08-05 2015-08-05 Carrier molecule for antigens WO2016020413A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
CN201580053997.XA CN108064174A (zh) 2014-08-05 2015-08-05 抗原的载体分子
AU2015299008A AU2015299008B2 (en) 2014-08-05 2015-08-05 Carrier molecule for antigens
SG11201700673RA SG11201700673RA (en) 2014-08-05 2015-08-05 Carrier molecule for antigens
KR1020177005840A KR20170039709A (ko) 2014-08-05 2015-08-05 항원에 대한 담체 분자
US15/501,474 US10245310B2 (en) 2014-08-05 2015-08-05 Carrier molecule for antigens
CA2957118A CA2957118A1 (en) 2014-08-05 2015-08-05 Carrier molecule for antigens
MX2017001638A MX2017001638A (es) 2014-08-05 2015-08-05 Molecula portadora para antigenos.
BR112017002183-8A BR112017002183A2 (pt) 2014-08-05 2015-08-05 conjugado, composição farmacêutica, e, método para aumentar uma resposta imune em um mamífero
JP2017506294A JP2017523985A (ja) 2014-08-05 2015-08-05 抗原用担体分子
EP15744610.5A EP3177324A1 (en) 2014-08-05 2015-08-05 Carrier molecule for antigens
EA201790239A EA201790239A1 (ru) 2014-08-05 2015-08-05 Молекула-носитель для антигенов
IL250238A IL250238A0 (en) 2014-08-05 2017-01-23 A carrier molecule for antigens
ZA2017/00824A ZA201700824B (en) 2014-08-05 2017-02-02 Carrier molecule for antigens

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP14179945.2 2014-08-05
EP14179945 2014-08-05

Publications (1)

Publication Number Publication Date
WO2016020413A1 true WO2016020413A1 (en) 2016-02-11

Family

ID=51298571

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/068012 WO2016020413A1 (en) 2014-08-05 2015-08-05 Carrier molecule for antigens

Country Status (16)

Country Link
US (1) US10245310B2 (zh)
EP (1) EP3177324A1 (zh)
JP (1) JP2017523985A (zh)
KR (1) KR20170039709A (zh)
CN (1) CN108064174A (zh)
AR (1) AR101455A1 (zh)
AU (1) AU2015299008B2 (zh)
BE (1) BE1022792B1 (zh)
BR (1) BR112017002183A2 (zh)
CA (1) CA2957118A1 (zh)
EA (1) EA201790239A1 (zh)
IL (1) IL250238A0 (zh)
MX (1) MX2017001638A (zh)
SG (1) SG11201700673RA (zh)
WO (1) WO2016020413A1 (zh)
ZA (1) ZA201700824B (zh)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018104889A1 (en) 2016-12-06 2018-06-14 Glaxosmithkline Biologicals Sa Purification process for capsular polysaccharide
WO2018229708A1 (en) 2017-06-16 2018-12-20 Glaxosmithkline Biologicals Sa Method of treatment
WO2020010000A1 (en) * 2018-07-04 2020-01-09 Sutrovax, Inc. Improved methods for the preparation of immunogenic conjugates
WO2020009993A1 (en) * 2018-07-04 2020-01-09 Sutrovax, Inc. Improvements in immunogenic conjugates
WO2020205584A1 (en) * 2019-04-02 2020-10-08 Vaxcyte, Inc. Optimized cell-free synthesis of invasion plasmid antigen b and related compositions and methods of use
WO2021099982A1 (en) 2019-11-22 2021-05-27 Glaxosmithkline Biologicals Sa Dosage and administration of a bacterial saccharide glycoconjugate vaccine
WO2021250625A2 (en) 2020-06-12 2021-12-16 Glaxosmithkline Biologicals Sa Gbs ferritin nanoparticles
WO2021250626A3 (en) * 2020-06-12 2022-02-24 Glaxosmithkline Biologicals Sa Dock tag system
US11951165B2 (en) 2016-12-30 2024-04-09 Vaxcyte, Inc. Conjugated vaccine carrier proteins
WO2024083958A1 (en) * 2022-10-21 2024-04-25 Glaxosmithkline Biologicals Sa Polypeptide scaffold

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2611960C (en) * 2005-06-27 2015-05-05 Glaxosmithkline Biologicals S.A. Immunogenic compositions comprising n.meningitidis capsular saccharide conjugates
WO2019217665A1 (en) * 2018-05-09 2019-11-14 West Virginia University Iron-acquisition receptor peptide administration for vaccination against pseudomonas aeruginosa
CN108840914B (zh) * 2018-08-13 2022-07-01 内蒙古民族大学 一种具有免疫原性的多肽、其抗体的制备方法以及用途
CN112979797B (zh) * 2021-04-28 2022-04-01 中国检验检疫科学研究院 一种抗赤羽病病毒单克隆抗体及其制备方法与应用

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006069200A2 (en) * 2004-12-22 2006-06-29 Novartis Vaccines And Diagnostics Inc. Group b streptococcus
WO2006078318A2 (en) * 2004-07-29 2006-07-27 Novartis Vaccines And Diagnostics Inc. Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
WO2009027768A2 (en) * 2006-07-26 2009-03-05 Novartis Ag Immunogenic compositions for gram positive bacteria
WO2009101403A1 (en) * 2008-02-11 2009-08-20 Novartis Ag Hybrid polypeptides comprising gbs-80 and spb1 proteins of streptococcus
WO2010079464A1 (en) * 2009-01-12 2010-07-15 Novartis Ag Cna_b domain antigens in vaccines against gram positive bacteria
GB2478203A (en) * 2010-02-26 2011-08-31 Novartis Ag Polypeptides and compositions for the treatment of Streptococcus agalactiae infection
WO2011121576A2 (en) * 2010-04-01 2011-10-06 Novartis Ag Immunogenic proteins and compositions
WO2013030783A1 (en) * 2011-08-30 2013-03-07 Novartis Ag Immunogenic proteins and compositions
WO2013124473A1 (en) * 2012-02-24 2013-08-29 Novartis Ag Pilus proteins and compositions

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0502095D0 (en) * 2005-02-01 2005-03-09 Chiron Srl Conjugation of streptococcal capsular saccharides
GB0605757D0 (en) * 2006-03-22 2006-05-03 Chiron Srl Separation of conjugated and unconjugated components

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006078318A2 (en) * 2004-07-29 2006-07-27 Novartis Vaccines And Diagnostics Inc. Immunogenic compositions for gram positive bacteria such as streptococcus agalactiae
WO2006069200A2 (en) * 2004-12-22 2006-06-29 Novartis Vaccines And Diagnostics Inc. Group b streptococcus
WO2009027768A2 (en) * 2006-07-26 2009-03-05 Novartis Ag Immunogenic compositions for gram positive bacteria
WO2009101403A1 (en) * 2008-02-11 2009-08-20 Novartis Ag Hybrid polypeptides comprising gbs-80 and spb1 proteins of streptococcus
WO2010079464A1 (en) * 2009-01-12 2010-07-15 Novartis Ag Cna_b domain antigens in vaccines against gram positive bacteria
GB2478203A (en) * 2010-02-26 2011-08-31 Novartis Ag Polypeptides and compositions for the treatment of Streptococcus agalactiae infection
WO2011121576A2 (en) * 2010-04-01 2011-10-06 Novartis Ag Immunogenic proteins and compositions
WO2013030783A1 (en) * 2011-08-30 2013-03-07 Novartis Ag Immunogenic proteins and compositions
WO2013124473A1 (en) * 2012-02-24 2013-08-29 Novartis Ag Pilus proteins and compositions

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CHENG QI ET AL: "Antibody against surface-bound C5a peptidase is opsonic and initiates macrophage killing of group B streptococci", INFECTION AND IMMUNITY, vol. 69, no. 4, April 2001 (2001-04-01), pages 2302 - 2308, XP002336337, ISSN: 0019-9567, DOI: 10.1128/IAI.69.4.2302-2308.2001 *
G. LINDAHL ET AL: "Surface Proteins of Streptococcus agalactiae and Related Proteins in Other Bacterial Pathogens", CLINICAL MICROBIOLOGY REVIEWS, vol. 18, no. 1, 2005, pages 102 - 127, XP055166916, ISSN: 0893-8512, DOI: 10.1128/CMR.18.1.102-127.2005 *
K. FLUEGGE ET AL: "Identification and immunoreactivity of proteins released from Streptococcus agalactiae", EUROPEAN JOURNAL OF CLINICAL MICROBIOLOGY & INFECTIOUS DISEASES, vol. 23, no. 11, November 2004 (2004-11-01), pages 818 - 824, XP055129256, ISSN: 0934-9723, DOI: 10.1007/s10096-004-1229-y *
LAWRENCE C. PAOLETTI ET AL: "Vaccines to prevent neonatal GBS infection", SEMINARS IN NEONATOLOGY, vol. 7, no. 4, August 2002 (2002-08-01), pages 315 - 323, XP055166924, ISSN: 1084-2756, DOI: 10.1053/siny.2002.0114 *
MADOFF L C ET AL: "MATERNAL IMMUNIZATION OF MICE WITH GROUP B STREPTOCOCCAL TYPE III POLYSACCHARIDE-BETA C PROTEIN CONJUGATE ELICITS PROTECTIVE ANTIBODY TO MULTIPLE SEROTYPES", JOURNAL OF CLINICAL INVESTIGATION, vol. 94, July 1994 (1994-07-01), pages 286 - 292, XP002923461, ISSN: 0021-9738, DOI: 10.1172/JCI117319 *
MONIKA BRZYCHCZY-WLOCH ET AL: "Identification of high immunoreactive proteins from Streptococcus agalactiae isolates recognized by human serum antibodies", FEMS MICROBIOLOGY LETTERS, vol. 349, 24 October 2013 (2013-10-24), pages 61 - 70, XP055129245, ISSN: 0378-1097, DOI: 10.1111/1574-6968.12292 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018104889A1 (en) 2016-12-06 2018-06-14 Glaxosmithkline Biologicals Sa Purification process for capsular polysaccharide
US11951165B2 (en) 2016-12-30 2024-04-09 Vaxcyte, Inc. Conjugated vaccine carrier proteins
WO2018229708A1 (en) 2017-06-16 2018-12-20 Glaxosmithkline Biologicals Sa Method of treatment
WO2020010000A1 (en) * 2018-07-04 2020-01-09 Sutrovax, Inc. Improved methods for the preparation of immunogenic conjugates
WO2020009993A1 (en) * 2018-07-04 2020-01-09 Sutrovax, Inc. Improvements in immunogenic conjugates
WO2020205584A1 (en) * 2019-04-02 2020-10-08 Vaxcyte, Inc. Optimized cell-free synthesis of invasion plasmid antigen b and related compositions and methods of use
WO2021099982A1 (en) 2019-11-22 2021-05-27 Glaxosmithkline Biologicals Sa Dosage and administration of a bacterial saccharide glycoconjugate vaccine
WO2021250625A2 (en) 2020-06-12 2021-12-16 Glaxosmithkline Biologicals Sa Gbs ferritin nanoparticles
WO2021250626A3 (en) * 2020-06-12 2022-02-24 Glaxosmithkline Biologicals Sa Dock tag system
WO2024083958A1 (en) * 2022-10-21 2024-04-25 Glaxosmithkline Biologicals Sa Polypeptide scaffold

Also Published As

Publication number Publication date
KR20170039709A (ko) 2017-04-11
EP3177324A1 (en) 2017-06-14
JP2017523985A (ja) 2017-08-24
IL250238A0 (en) 2017-03-30
ZA201700824B (en) 2019-07-31
AU2015299008B2 (en) 2018-08-02
EA201790239A1 (ru) 2017-07-31
AU2015299008A1 (en) 2017-02-23
BR112017002183A2 (pt) 2018-01-16
CN108064174A (zh) 2018-05-22
MX2017001638A (es) 2017-04-27
BE1022792B1 (fr) 2016-09-06
SG11201700673RA (en) 2017-02-27
AR101455A1 (es) 2016-12-21
BE1022792A1 (fr) 2016-09-06
CA2957118A1 (en) 2016-02-11
US10245310B2 (en) 2019-04-02
US20170224803A1 (en) 2017-08-10

Similar Documents

Publication Publication Date Title
US10245310B2 (en) Carrier molecule for antigens
AU2012335208B2 (en) Carrier molecule comprising a spr0096 and a spr2021 antigen
US10188719B2 (en) Conjugation of Streptococcal capsular saccharides
US9173954B2 (en) Polysaccharide immunogens conjugated to E. coli carrier proteins
US10736959B2 (en) Conjugation of Staphylococcus aureus type 5 and type 8 capsular polysaccharides
EP2227248B1 (en) Adjuvanted glucans

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15744610

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 250238

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2957118

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2017506294

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2017/001638

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015744610

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201790239

Country of ref document: EA

Ref document number: 2015744610

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017002183

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2015299008

Country of ref document: AU

Date of ref document: 20150805

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20177005840

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112017002183

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20170202